LayerRx Mapping ID
100
Slot System
Featured Buckets
Featured Buckets Admin

10 recommendations for the Cancer Moonshot

Article Type
Changed
Thu, 03/28/2019 - 15:03
Display Headline
10 recommendations for the Cancer Moonshot

Responding to the Cancer Moonshot initiative, a panel of scientists, clinicians, patient advocates, and industry representatives has issued 10 recommendations for accelerating cancer research in an article published in Science.

The recommendations address:

• Development of a patient engagement network.

• Precise cataloging of tumor molecular changes.

• Analysis of samples already available from patients who have received the standard of care.

 

©Tashatuvango/Thinkstock

• Improvements for data sharing, access, and analysis.

• Development of models to understand how childhood cancers develop.

• Research to describe how fusion oncoproteins drive cancer development.

• Creation of a cancer immunotherapy clinical trials network.

• Systematic efforts to gather information on patient-reported outcomes.

• Implementation of evidence-based approaches to prevention.

The panel’s recommendations were presented to the National Cancer Advisory Board, the adviser to the National Cancer Institute. The ability to conduct research stemming from the panel’s recommendations will depend on whether, and how much, funding is approved by Congress.

Read the article here: http://science.sciencemag.org/content/early/2016/09/07/science.aai7862.full.

Publications
Topics

Responding to the Cancer Moonshot initiative, a panel of scientists, clinicians, patient advocates, and industry representatives has issued 10 recommendations for accelerating cancer research in an article published in Science.

The recommendations address:

• Development of a patient engagement network.

• Precise cataloging of tumor molecular changes.

• Analysis of samples already available from patients who have received the standard of care.

 

©Tashatuvango/Thinkstock

• Improvements for data sharing, access, and analysis.

• Development of models to understand how childhood cancers develop.

• Research to describe how fusion oncoproteins drive cancer development.

• Creation of a cancer immunotherapy clinical trials network.

• Systematic efforts to gather information on patient-reported outcomes.

• Implementation of evidence-based approaches to prevention.

The panel’s recommendations were presented to the National Cancer Advisory Board, the adviser to the National Cancer Institute. The ability to conduct research stemming from the panel’s recommendations will depend on whether, and how much, funding is approved by Congress.

Read the article here: http://science.sciencemag.org/content/early/2016/09/07/science.aai7862.full.

Responding to the Cancer Moonshot initiative, a panel of scientists, clinicians, patient advocates, and industry representatives has issued 10 recommendations for accelerating cancer research in an article published in Science.

The recommendations address:

• Development of a patient engagement network.

• Precise cataloging of tumor molecular changes.

• Analysis of samples already available from patients who have received the standard of care.

 

©Tashatuvango/Thinkstock

• Improvements for data sharing, access, and analysis.

• Development of models to understand how childhood cancers develop.

• Research to describe how fusion oncoproteins drive cancer development.

• Creation of a cancer immunotherapy clinical trials network.

• Systematic efforts to gather information on patient-reported outcomes.

• Implementation of evidence-based approaches to prevention.

The panel’s recommendations were presented to the National Cancer Advisory Board, the adviser to the National Cancer Institute. The ability to conduct research stemming from the panel’s recommendations will depend on whether, and how much, funding is approved by Congress.

Read the article here: http://science.sciencemag.org/content/early/2016/09/07/science.aai7862.full.

Publications
Publications
Topics
Article Type
Display Headline
10 recommendations for the Cancer Moonshot
Display Headline
10 recommendations for the Cancer Moonshot
Disallow All Ads

Gene profile predicts RCC response to nivolumab

Article Type
Changed
Fri, 01/04/2019 - 13:19
Display Headline
Gene profile predicts RCC response to nivolumab

Many patients with advanced renal cell carcinoma have tumors that do not respond to immune checkpoint inhibitors targeted against the programmed death-1 (PD-1) pathway, despite expression of the target PD ligand 1 (PD-L1) on their tumors. Now investigators think they know why, and hope to use the information to predict which patients are likely to benefit and identify potential new therapies or combinations.

A study of renal cell carcinoma (RCC) samples from tumors with both good and poor clinical responses to treatment with the anti–PD-1 agent nivolumab (Opdivo) showed that a tumor gene–expression profile tipped more toward genes for controlling metabolic functions rather than immune functions was associated with a lack of response to anti-PD-1 therapy, reported Suzanne L. Topalian, MD, and her colleagues from Johns Hopkins University and the Sidney Kimmel Comprehensive Cancer Center, both in Baltimore.

 

Dr. Suzanne Topalian

“These findings suggest that tumor cell–intrinsic metabolic factors may contribute to treatment resistance in RCC, thus serving as predictive markers for treatment outcomes and potential new targets for combination therapy regimens with anti–PD-1,” they wrote in a study published online in Cancer Immunology Research.

The investigators obtained tumor samples from 13 patients with unresectable metastatic RCC treated in one of four clinical trials. They used radiographic staging to classify each patient as either a responder or nonresponder to anti–PD-1 therapy according to RECIST (Response Evaluation Criteria in Solid Tumors). The samples were evaluated with whole genome microarray and multiplex quantitative reverse-transcription polymerase chain reaction (qRT-PCR) profiling and analysis, and the results were compared with those from eight renal cell carcinoma cell lines.

They looked for expression of nearly 30,000 gene targets in samples from responders and nonresponders and found a pattern of differential expression of genes encoding for metabolic pathways and immune functions.

Specifically, they found that the expression of genes involved in metabolic and solute transport functions (for example, UGT1A) were associated with poor response to nivolumab, whereas overexpression of genes for immune markers involved in T-cell differentiation (BACH2) and leukocyte migration (CCL3) were associated with a good response.

The investigators acknowledge that the study was retrospective and limited by the analysis of only a small number of tumor samples but suggest that their findings point the way to further investigations in larger groups of patients with RCC tumors, including those both positive and negative for PD-L1 expression.

“The general approach to identifying biomarkers of clinical response to PD-1–targeted therapies has so far focused on immunologic factors in the [tumor microenvironment]. However, a deeper level of investigation may be warranted for individual tumor types, and intersections of tumor cell–intrinsic factors with immunologic factors may be particularly revealing,” they wrote.

The study was supported by research grants from the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Bristol-Myers Squibb, the National Cancer Institute, and Stand Up To Cancer. Dr. Topalian has served as a consultant/advisory board member for Five Prime Therapeutics, MedImmune, Merck, and Pfizer, and has an ownership interest in Bristol-Myers Squibb, Five Prime Therapeutics,and Potenza Therapeutics. Other coauthors reported similar potential conflicts of interest.

Publications
Topics

Many patients with advanced renal cell carcinoma have tumors that do not respond to immune checkpoint inhibitors targeted against the programmed death-1 (PD-1) pathway, despite expression of the target PD ligand 1 (PD-L1) on their tumors. Now investigators think they know why, and hope to use the information to predict which patients are likely to benefit and identify potential new therapies or combinations.

A study of renal cell carcinoma (RCC) samples from tumors with both good and poor clinical responses to treatment with the anti–PD-1 agent nivolumab (Opdivo) showed that a tumor gene–expression profile tipped more toward genes for controlling metabolic functions rather than immune functions was associated with a lack of response to anti-PD-1 therapy, reported Suzanne L. Topalian, MD, and her colleagues from Johns Hopkins University and the Sidney Kimmel Comprehensive Cancer Center, both in Baltimore.

 

Dr. Suzanne Topalian

“These findings suggest that tumor cell–intrinsic metabolic factors may contribute to treatment resistance in RCC, thus serving as predictive markers for treatment outcomes and potential new targets for combination therapy regimens with anti–PD-1,” they wrote in a study published online in Cancer Immunology Research.

The investigators obtained tumor samples from 13 patients with unresectable metastatic RCC treated in one of four clinical trials. They used radiographic staging to classify each patient as either a responder or nonresponder to anti–PD-1 therapy according to RECIST (Response Evaluation Criteria in Solid Tumors). The samples were evaluated with whole genome microarray and multiplex quantitative reverse-transcription polymerase chain reaction (qRT-PCR) profiling and analysis, and the results were compared with those from eight renal cell carcinoma cell lines.

They looked for expression of nearly 30,000 gene targets in samples from responders and nonresponders and found a pattern of differential expression of genes encoding for metabolic pathways and immune functions.

Specifically, they found that the expression of genes involved in metabolic and solute transport functions (for example, UGT1A) were associated with poor response to nivolumab, whereas overexpression of genes for immune markers involved in T-cell differentiation (BACH2) and leukocyte migration (CCL3) were associated with a good response.

The investigators acknowledge that the study was retrospective and limited by the analysis of only a small number of tumor samples but suggest that their findings point the way to further investigations in larger groups of patients with RCC tumors, including those both positive and negative for PD-L1 expression.

“The general approach to identifying biomarkers of clinical response to PD-1–targeted therapies has so far focused on immunologic factors in the [tumor microenvironment]. However, a deeper level of investigation may be warranted for individual tumor types, and intersections of tumor cell–intrinsic factors with immunologic factors may be particularly revealing,” they wrote.

The study was supported by research grants from the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Bristol-Myers Squibb, the National Cancer Institute, and Stand Up To Cancer. Dr. Topalian has served as a consultant/advisory board member for Five Prime Therapeutics, MedImmune, Merck, and Pfizer, and has an ownership interest in Bristol-Myers Squibb, Five Prime Therapeutics,and Potenza Therapeutics. Other coauthors reported similar potential conflicts of interest.

Many patients with advanced renal cell carcinoma have tumors that do not respond to immune checkpoint inhibitors targeted against the programmed death-1 (PD-1) pathway, despite expression of the target PD ligand 1 (PD-L1) on their tumors. Now investigators think they know why, and hope to use the information to predict which patients are likely to benefit and identify potential new therapies or combinations.

A study of renal cell carcinoma (RCC) samples from tumors with both good and poor clinical responses to treatment with the anti–PD-1 agent nivolumab (Opdivo) showed that a tumor gene–expression profile tipped more toward genes for controlling metabolic functions rather than immune functions was associated with a lack of response to anti-PD-1 therapy, reported Suzanne L. Topalian, MD, and her colleagues from Johns Hopkins University and the Sidney Kimmel Comprehensive Cancer Center, both in Baltimore.

 

Dr. Suzanne Topalian

“These findings suggest that tumor cell–intrinsic metabolic factors may contribute to treatment resistance in RCC, thus serving as predictive markers for treatment outcomes and potential new targets for combination therapy regimens with anti–PD-1,” they wrote in a study published online in Cancer Immunology Research.

The investigators obtained tumor samples from 13 patients with unresectable metastatic RCC treated in one of four clinical trials. They used radiographic staging to classify each patient as either a responder or nonresponder to anti–PD-1 therapy according to RECIST (Response Evaluation Criteria in Solid Tumors). The samples were evaluated with whole genome microarray and multiplex quantitative reverse-transcription polymerase chain reaction (qRT-PCR) profiling and analysis, and the results were compared with those from eight renal cell carcinoma cell lines.

They looked for expression of nearly 30,000 gene targets in samples from responders and nonresponders and found a pattern of differential expression of genes encoding for metabolic pathways and immune functions.

Specifically, they found that the expression of genes involved in metabolic and solute transport functions (for example, UGT1A) were associated with poor response to nivolumab, whereas overexpression of genes for immune markers involved in T-cell differentiation (BACH2) and leukocyte migration (CCL3) were associated with a good response.

The investigators acknowledge that the study was retrospective and limited by the analysis of only a small number of tumor samples but suggest that their findings point the way to further investigations in larger groups of patients with RCC tumors, including those both positive and negative for PD-L1 expression.

“The general approach to identifying biomarkers of clinical response to PD-1–targeted therapies has so far focused on immunologic factors in the [tumor microenvironment]. However, a deeper level of investigation may be warranted for individual tumor types, and intersections of tumor cell–intrinsic factors with immunologic factors may be particularly revealing,” they wrote.

The study was supported by research grants from the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Bristol-Myers Squibb, the National Cancer Institute, and Stand Up To Cancer. Dr. Topalian has served as a consultant/advisory board member for Five Prime Therapeutics, MedImmune, Merck, and Pfizer, and has an ownership interest in Bristol-Myers Squibb, Five Prime Therapeutics,and Potenza Therapeutics. Other coauthors reported similar potential conflicts of interest.

Publications
Publications
Topics
Article Type
Display Headline
Gene profile predicts RCC response to nivolumab
Display Headline
Gene profile predicts RCC response to nivolumab
Article Source

FROM CANCER IMMUNOLOGY RESEARCH

Disallow All Ads
Vitals

Key clinical point: Many renal cell carcinoma tumors do not respond to therapy with an anti-PD-1 agent, despite being positive for the PD-L1 target.

Major finding: A gene expression profile favoring genes associated with metabolic and solute transport functions was associated with poor response to nivolumab.

Data source: Retrospective study of tumor gene expression in 13 patients with advanced RCC and 8 RCC cell lines.

Disclosures: The study was supported by research grants from the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore; Bristol-Myers Squibb; the National Cancer Institute; and Stand Up To Cancer. Dr. Topalian has served as a consultant/advisory board member for Five Prime Therapeutics, MedImmune, Merck, and Pfizer, and has an ownership interest in Bristol-Myers Squibb, Five Prime Therapeutics,and Potenza Therapeutics. Other coauthors reported similar potential conflicts of interest.

The Cost of Oncology Drugs: A Pharmacy Perspective, Part 2

Article Type
Changed
Tue, 04/24/2018 - 16:23
Display Headline
The Cost of Oncology Drugs: A Pharmacy Perspective, Part 2
Guidance documents, the Pharmacy Benefits Management Services, and the VA National Formulary help clinicians and pharmacists decide whether to administer high-cost cancer treatments.

The Cost of Oncology Drugs: A Pharmacy Perspective, Part 1, appeared in the Federal Practitioner February 2016 special issue “Best Practices in Hematology and Oncology” and can be accessed here.

Health care costs are the fastest growing financial segment of the U.S. economy. The cost of medications, especially those for treating cancer, is the leading cause of increased health care spending.1 Until recently, the discussion of the high costs of cancer treatment was rarely made public.

Part 1 of this article focused on the emerging discussion of the financial impact of high-cost drugs in the U.S. Part 2 will focus on the drivers of increasing oncology drug costs and the challenges high-cost medications pose for the VA. The article also will review the role of the VA Pharmacy Benefits Management Service (PBM) in evaluating new oncology agents. Also presented are the clinical guidance tools designed to aid the clinician in the cost-effective use of these agents and results of a nationwide survey of VA oncology pharmacists regarding the use of cost-containment strategies.

 

Cost Drivers

Many factors are driving increased oncology drug costs within the VA. Although the cost of individual drugs has the largest impact on the accelerating cost of treating each patient, other clinical and social factors may play a role.

Increasing Cost of Individual Drugs

Drug pricing is not announced until after FDA approval. Oncology drugs at the high end of the cost spectrum are rarely curative and often add only weeks or months to overall survival (OS), the gold standard. Current clinical trial design often uses progression free survival (PFS) as the primary endpoint, which makes the use of traditional pharmacoeconomic determinations of value difficult. In addition, many new drugs are first in class and/or have narrow indications that preclude competition from other drugs. Although addressing the issue of the market price for drugs seems to be one that is not controllable, there is increasing demand for drug pricing reform.2

Many believe drug prices should be linked directly to clinical benefit. In a recent article, Goldstein and colleagues proposed establishing a value-based price for necitumumab based on clinical benefit, prior to FDA approval.3 When this analysis was done, necitumumab was pending FDA approval in combination with cisplatin and gemcitabine for the treatment of squamous carcinoma of the lung. Using clinical data from the SQUIRE trial on which FDA approval was based, the addition of necitumumab to the chemotherapy regimen led to an incremental survival benefit of 0.15 life-years and 0.11 quality-adjusted life-years (QALY).4 Using a Markov model to evaluate cost-effectiveness, these authors established that the price of necitumumab should be from $563 to $1,309 per cycle. Necitumumab was approved by the FDA on November 24, 2015, with the VA acquisition cost, as of May 2016, at $6,100 per cycle.

Lack of Generic Products

The approval of generic alternatives for targeted oncology agents should reduce the cost of treating oncology patients. However, since imatinib was approved in May 2001, no single targeted agent had become available as a generic until February 1, 2016, when generic imatinib was made available in the U.S. following approval by the FDA. Currently, generic imatinib is not used in the VA due to lack of Federal Supply Schedule (FSS) contract pricing, which leads to a generic cost that is much higher than the brand-name drug, Gleevec ($6,127 per month vs $9,472 per month for the generic). The reality is that many older agents have steadily increased in price, outpacing inflation (Table 1).5

Aging U.S. Population

Advancing age is the most common risk factor for cancer, leading to an increase in the incidence and treatment of cancer. Because many newer agents are considered easier to tolerate than are traditional cytotoxic chemotherapy, clinicians have become more comfortable treating elderly patients, and geriatric oncology has become an established subspecialty within oncology.

Changing Treatment Paradigms

The use of targeted therapies is changing the paradigm from the acute treatment of cancer to chronic cancer management. Most targeted therapies are continued until disease progression or toxicity, leading to chronic, open-ended treatment. This approach is in contrast to older treatment approaches such as chemotherapy, which is often given for a limited duration followed by observation. When successful, chronic treatment with targeted agents can lead to unanticipated high costs. The following current cases at the VA San Diego Healthcare System illustrate this point:

  • Renal cell carcinoma: 68-year-old man diagnosed in 2005 with a recurrence in 2012

- High-dose interleukin-2 (2 cycles); sunitinib (3.3 years); pazopanib (2 months); everolimus (2 months); sorafenib (3 months); axitinib (7 months)

- Now contemplating anti-PD-1, pending emerging data. Total cost to date ~ $324,300.
 

 

  • Cutaneous T-cell lymphoma: 68-year-old man started romidepsin September 22, 2010
- Received 108 doses through May 2016. Total cost to date~ $652,970.

The rate of FDA approval for oncology drugs has been accelerating rapidly in the past 15 years. Sequential therapies beyond second-line therapy are common as more agents become available. Table 2 shows FDA approval for all cancer drugs by decade.

As researchers continue to better understand the many pathways involved with the development and progression of cancer, they are beginning to combine multiple targeted agents to augment response rates, prolong survival, and reduce the potential for resistance. Recent combination regimens approved by the FDA include dabrafenib plus trametinib (January 2014), and ipilimumab plus nivolumab (October 2015), both for the treatment of melanoma. In November 2015, ixazomib was FDA approved to be used in combination with lenalidomide for multiple myeloma. Many more combination regimens are currently in clinical trials, and more combinations are expected to receive FDA approval. It is easy to see how the combination of multiple expensive agents with the prospect of prolonged therapy has the potential to increase the cost of many regimens to well over $100,000 per year.

Maintenance therapy is used to prolong PFS for patients receiving an excellent response to primary therapy. For example, VA costs for maintenance regimens include lenalidomide 10 mg daily: $8,314 for 28 days equals $216,177 for 2 years; bortezomib 1.3 mg/m2 (2.6 mg) q: 2 weeks equals $60,730 for 2 years (includes waste as bortezomib 3.5-mg vials do not a contain preservative and must be discarded within 8 hours of preparation); and rituximab 800 mg q: 2 months equals $47,635 for 2 years.

Until recently, immunotherapy for cancer was limited to melanoma and renal cell carcinoma using interleukin-2 (aldesleukin) and interferon alfa. However, the immergence of new immunotherapies, such as anti-PD-1 and anti-CTLA-4 monoclonal antibodies, have expanded the role of immunotherapy to many other, more common, malignancies, such as lung cancer, breast cancer, prostate cancer, head and neck cancer, and many more.

Most randomized clinical trials study drugs as second- or occasionally third-line therapy. However, many patients continue to be treated beyond the third-line setting, often without evidence-based data to support potential benefit. Patients often place value on treatments unlikely to work so as not to give up hope. These “hopeful gambles,” even with the potential of significant toxicity and decreased quality of life (QOL), are common in cancer treatment.6 In addition, oncologists often overestimate the clinical benefit when considering additional therapy in this setting.7

Influx of New Patients

Outside the VHA setting, the financial burden of cancer treatment has led to an influx of new patients transferring care to the VHA to reduce out-of-pocket expenses. Because private insurance copays for oral agents are increasing, many reaching 20% to 30%, out-of-pocket expenses for medications can reach several thousand dollars per month. Patients often change insurance plans due to changing jobs or to decrease cost, or employers may change plans to save money, which may significantly alter or discontinue coverage. Patients often request that the VA provide medication while continuing to see only their private oncologist. This practice should be discouraged because the VA, without clinical involvement, may supply drugs for inappropriate indications. In addition, VA providers writing prescriptions for medications without personally following patients may be liable for poor outcomes.

VA PBM Services

Prior to 1995, the VA was a much criticized and poorly performing health care system that had experienced significant budget cuts, forcing many veterans to seek care outside the VA. Then beginning in 1995, a remarkable transformation occurred, which modernized and improved the VA into a system that consistently outperforms the private sector in quality of care, patient safety, patient satisfaction, all at a lower cost.8 The story of the VA’s transformation has been well chronicled by Phillip Longman.9

Under the direction of VA Under Secretary for Health Kenneth Kizer, MD, MPH, VA established PBM Clinical Services to develop and maintain the National Drug Formulary, create clinical guidance documents, and manage drug costs and utilization. A recent article by Heron and Geraci examined the functions and role of the VA PBM in controlling oncology drug costs.10 The following is a brief review of several documents and VA PBM responsibilities as reviewed by Heron and Geraci.

VA National Formulary

Prior to the establishment of the VA National Formulary in 1995, each VA maintained its own formulary, which led to extreme variability in drug access across the country. When a patient accessed care at different VAMCs, it was common for the patient’s medications to be changed based on the specific facility formulary. This practice led to many potential problems, such as lack of clinical benefit and potentially increased or new toxicities, and led to extra hospital visits for monitoring and adjustment of medications.

 

 

In contrast, the VA National Formulary now offers a uniform pharmacy benefit to all veterans by reducing variation in access to drugs. In addition, using preferred agents in each drug class provides VA with additional leverage when contracting with drug suppliers to reduce prices across the entire VA system.

Many oncology agents are not included on the VA National Formulary due to cost and the potential for off-label use. However, the formulary status of oncology agents in no way limits access or the availability of any oncology drug for appropriate patients. In fact, nonformulary approval requests work as a mechanism for review to ensure that these agents are used properly in the subset of patients who are most likely to benefit.

The PBM assesses all new oncology drugs for value and potential use within the VA, as well as cost impact. Following this assessment, various clinical guidance documents may be developed that are intended to guide clinicians in the proper use of medications for veterans. All documents prepared by the PBM undergo an extensive peer review by the Medical Advisory Panel and other experts in the field.

 

Drug Monographs

A drug monograph is a comprehensive, evidence-based drug review that summarizes efficacy and safety based on clinical trial data published in peer-reviewed journals, abstracts, and/or FDA Medical Review transcripts. Cost-effectiveness analysis is included if available.

Criteria for Use

Criteria for Use (CFU) are developed for drugs considered to be at high risk for inappropriate use or with safety concerns. The purpose of the CFU is to select patients most likely to benefit from these agents by using clinical criteria, which may qualify or eliminate a patient for treatment. National CFUs are available on the national PBM website. Local CFUs are often written and shared among oncology pharmacists via the VA oncology pharmacist listserv.

Abbreviated Reviews

Similar to drug monographs, abbreviated reviews are much shorter and focus on the relevant clinical sections of the drug monograph necessary for clinical or formulary decision making.

National Acquisition Center

The National Acquisition Center (NAC) is the pharmaceutical contracting mechanism for the VA and works closely with the PBM.5 The NAC pursues significant drug price reductions for the VA based on many strategies. Public Law 102-585 ensures that certain government agencies, including the VA, receive special discounts on pharmaceuticals, which is at least a 24% discount from the nonfederal Average Manufacturer Price. This is known as the Federal Supply Schedule (FSS) and/or Big 4 pricing. In addition, bulk purchases and performance-based incentive agreements can lead to substantial local discounts. By working with specific drug distribution and warehouse contractors, the NAC assures ready access to drugs for VA patients. The NAC also allows for an efficient drug inventory process, thus reducing inventory management costs.

Guidance Documents

In 2012, the VA Oncology Field Advisory Committee (FAC) created the High Cost Oncology Drug Work Group to address the impact of high-cost oncology drugs within the VA.11 This work group was composed of VA oncologists and pharmacists whose efforts resulted in 5 guidance documents designed to reduce drug costs by optimizing therapy and reducing waste: (1) Dose Rounding in Oncology; (2) Oral Anticancer Drugs Dispensing and Monitoring; (3) Oncology Drug Table: Recommended Dispensing and Monitoring; (4) Chemotherapy Review Committee Process; and (5) Determining Clinical Benefit of High Cost Oncology Drugs. Reviews of 2 of these documents follows.

Determining Clinical Benefit of High Cost Oncology Drugs provides a decision tool to aid members of the oncology health care team in optimizing patient outcomes while attempting to obtain the greatest value from innovative therapies. When a high-cost or off-label request is made for a particular patient, using this process encourages thoughtful and evidence-based use of the drug by considering all clinical evidence in addition to the FDA-approved indication. Finally, a drug’s safety profile in relation to the indication, therapeutic goal, and specific patient characteristics and desires are integrated into a final decision to determine the appropriateness of the therapeutic intervention for the patient.

Oncology Drug Table: Recommended Dispensing and Monitoring contains a list of oral oncology drugs and includes recommendations for dispensing amount, adverse effects, laboratory monitoring, formulary status, approval requirements, and monthly cost of each agent based on the current NAC pricing.5 Cost awareness is critical when comparing alternative treatment options to minimize cost when treatments with similar benefits are considered. Most VA oncologists do not have easy access to the cost of various treatments and can be surprised about how expensive many common regimens cost. The costs listed in this document are updated about every 3 months.

 

 

Conclusion

Using newer, expensive targeted oncology agents in a cost-effective manner must be a proactive, collaborative, and multidisciplinary process. Pharmacists should not be solely responsible for monitoring and controlling high-cost treatments. Well-informed, evidence-based decisions are needed to ensure expensive agents are used in the subset of patients who are most likely to benefit. Clinical tools addressing value should be used to aid in appropriate and cost-effective treatment plans using drug monographs and CFUs, VHA Guidance on Determining Clinical Benefit of High Cost Oncology Drugs, and the Oral Chemotherapy Dispensing and Monitoring Reference, among other resources. Due to the subjective nature of value in medicine, agreeing on policy will have many challenges, such as how to place a value on various gains in overall survival, progression free survival, response rates, and QOL.

eAppendix

References

1. Bach PB. Limits on Medicare's ability to control rising spending on cancer drugs. N Engl J Med. 2009;360(6):626-633.

2. Kantarjian H, Steensma D, Rius Sanjuan J, Eishaug A, Light D. High cancer drug prices in the United States: reasons and proposed solutions. J Oncol Pract. 2014;10(4):e208-e211.

3. Goldstein DA, Chen Q, Ayer T, et al. Necitumumab in metastatic squamous cell lung cancer: establishing a value-based cost. JAMA Oncol. 2015;1(9):1293-1300.

4. Thatcher N, Hirsch FR, Luft AV, et al; SQUIRE Investigators. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015;16(7):763-774.

5. U.S. Department of Veterans Affairs, National Acquisition Center, Pharmaceutical Catalog Search. U.S. Department of Veterans Affairs, National Acquisition Center website. http://www1.va.gov/nac/index.cfm?template=Search_Pharmaceutical_Catalog. Updated June 13, 2016. Accessed June 13, 2016.

6. Lakdawalla DN, Romley JA, Sanchez Y, Maclean JR, Penrod JR, Philipson T. How cancer patients value hope and the implications for cost-effectiveness assessments of high-cost cancer therapies. Health Aff (Millwood). 2012;31(4):676-682.

7. Ubel PA, Berry SR, Nadler E, et al. In a survey, marked inconsistency in how oncologists judged value of high-cost cancer drugs in relation to gains in survival. Health Aff (Millwood). 2012;31(4):709-717.

8. Asch SM, McGlynn EA, Hogan MM, et al. Comparison of quality of care for patients in the Veterans Health Administration and patients in a national sample. Ann Intern Med. 2004;141(12):938-945. 9. Longman P. Best Care Anywhere: Why VA Health Care Would Work for Everyone. 3rd ed. San Francisco, CA: Berrett-Koehler Publishers; 2012. 10.  Heron BB, Geraci MC. Controlling the cost of oncology drugs within the VA: a national perspective. Fed Pract. 2015;32(suppl 1):18S-22S.

11.  U.S. Department of Veterans Affairs. Pharmacy Benefits Management Services Intranet, Documents and Lists. https://vaww.cmopnational.va.gov/cmop/PBM/Clinical%20Guidance/Forms/AllItems.aspx. Accessed May 19, 2016.

Article PDF
Author and Disclosure Information

Dr. Dahl is a clinical pharmacy specialist in oncology and director of the Oncology Pharmacy Program at the VA San Diego Healthcare System in California.

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 33(7)
Publications
Topics
Page Number
35-39
Legacy Keywords
oncology, medication, VA National Formulary, cancer treatment, imatinib
Sections
Author and Disclosure Information

Dr. Dahl is a clinical pharmacy specialist in oncology and director of the Oncology Pharmacy Program at the VA San Diego Healthcare System in California.

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Dr. Dahl is a clinical pharmacy specialist in oncology and director of the Oncology Pharmacy Program at the VA San Diego Healthcare System in California.

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Related Articles
Guidance documents, the Pharmacy Benefits Management Services, and the VA National Formulary help clinicians and pharmacists decide whether to administer high-cost cancer treatments.
Guidance documents, the Pharmacy Benefits Management Services, and the VA National Formulary help clinicians and pharmacists decide whether to administer high-cost cancer treatments.

The Cost of Oncology Drugs: A Pharmacy Perspective, Part 1, appeared in the Federal Practitioner February 2016 special issue “Best Practices in Hematology and Oncology” and can be accessed here.

Health care costs are the fastest growing financial segment of the U.S. economy. The cost of medications, especially those for treating cancer, is the leading cause of increased health care spending.1 Until recently, the discussion of the high costs of cancer treatment was rarely made public.

Part 1 of this article focused on the emerging discussion of the financial impact of high-cost drugs in the U.S. Part 2 will focus on the drivers of increasing oncology drug costs and the challenges high-cost medications pose for the VA. The article also will review the role of the VA Pharmacy Benefits Management Service (PBM) in evaluating new oncology agents. Also presented are the clinical guidance tools designed to aid the clinician in the cost-effective use of these agents and results of a nationwide survey of VA oncology pharmacists regarding the use of cost-containment strategies.

 

Cost Drivers

Many factors are driving increased oncology drug costs within the VA. Although the cost of individual drugs has the largest impact on the accelerating cost of treating each patient, other clinical and social factors may play a role.

Increasing Cost of Individual Drugs

Drug pricing is not announced until after FDA approval. Oncology drugs at the high end of the cost spectrum are rarely curative and often add only weeks or months to overall survival (OS), the gold standard. Current clinical trial design often uses progression free survival (PFS) as the primary endpoint, which makes the use of traditional pharmacoeconomic determinations of value difficult. In addition, many new drugs are first in class and/or have narrow indications that preclude competition from other drugs. Although addressing the issue of the market price for drugs seems to be one that is not controllable, there is increasing demand for drug pricing reform.2

Many believe drug prices should be linked directly to clinical benefit. In a recent article, Goldstein and colleagues proposed establishing a value-based price for necitumumab based on clinical benefit, prior to FDA approval.3 When this analysis was done, necitumumab was pending FDA approval in combination with cisplatin and gemcitabine for the treatment of squamous carcinoma of the lung. Using clinical data from the SQUIRE trial on which FDA approval was based, the addition of necitumumab to the chemotherapy regimen led to an incremental survival benefit of 0.15 life-years and 0.11 quality-adjusted life-years (QALY).4 Using a Markov model to evaluate cost-effectiveness, these authors established that the price of necitumumab should be from $563 to $1,309 per cycle. Necitumumab was approved by the FDA on November 24, 2015, with the VA acquisition cost, as of May 2016, at $6,100 per cycle.

Lack of Generic Products

The approval of generic alternatives for targeted oncology agents should reduce the cost of treating oncology patients. However, since imatinib was approved in May 2001, no single targeted agent had become available as a generic until February 1, 2016, when generic imatinib was made available in the U.S. following approval by the FDA. Currently, generic imatinib is not used in the VA due to lack of Federal Supply Schedule (FSS) contract pricing, which leads to a generic cost that is much higher than the brand-name drug, Gleevec ($6,127 per month vs $9,472 per month for the generic). The reality is that many older agents have steadily increased in price, outpacing inflation (Table 1).5

Aging U.S. Population

Advancing age is the most common risk factor for cancer, leading to an increase in the incidence and treatment of cancer. Because many newer agents are considered easier to tolerate than are traditional cytotoxic chemotherapy, clinicians have become more comfortable treating elderly patients, and geriatric oncology has become an established subspecialty within oncology.

Changing Treatment Paradigms

The use of targeted therapies is changing the paradigm from the acute treatment of cancer to chronic cancer management. Most targeted therapies are continued until disease progression or toxicity, leading to chronic, open-ended treatment. This approach is in contrast to older treatment approaches such as chemotherapy, which is often given for a limited duration followed by observation. When successful, chronic treatment with targeted agents can lead to unanticipated high costs. The following current cases at the VA San Diego Healthcare System illustrate this point:

  • Renal cell carcinoma: 68-year-old man diagnosed in 2005 with a recurrence in 2012

- High-dose interleukin-2 (2 cycles); sunitinib (3.3 years); pazopanib (2 months); everolimus (2 months); sorafenib (3 months); axitinib (7 months)

- Now contemplating anti-PD-1, pending emerging data. Total cost to date ~ $324,300.
 

 

  • Cutaneous T-cell lymphoma: 68-year-old man started romidepsin September 22, 2010
- Received 108 doses through May 2016. Total cost to date~ $652,970.

The rate of FDA approval for oncology drugs has been accelerating rapidly in the past 15 years. Sequential therapies beyond second-line therapy are common as more agents become available. Table 2 shows FDA approval for all cancer drugs by decade.

As researchers continue to better understand the many pathways involved with the development and progression of cancer, they are beginning to combine multiple targeted agents to augment response rates, prolong survival, and reduce the potential for resistance. Recent combination regimens approved by the FDA include dabrafenib plus trametinib (January 2014), and ipilimumab plus nivolumab (October 2015), both for the treatment of melanoma. In November 2015, ixazomib was FDA approved to be used in combination with lenalidomide for multiple myeloma. Many more combination regimens are currently in clinical trials, and more combinations are expected to receive FDA approval. It is easy to see how the combination of multiple expensive agents with the prospect of prolonged therapy has the potential to increase the cost of many regimens to well over $100,000 per year.

Maintenance therapy is used to prolong PFS for patients receiving an excellent response to primary therapy. For example, VA costs for maintenance regimens include lenalidomide 10 mg daily: $8,314 for 28 days equals $216,177 for 2 years; bortezomib 1.3 mg/m2 (2.6 mg) q: 2 weeks equals $60,730 for 2 years (includes waste as bortezomib 3.5-mg vials do not a contain preservative and must be discarded within 8 hours of preparation); and rituximab 800 mg q: 2 months equals $47,635 for 2 years.

Until recently, immunotherapy for cancer was limited to melanoma and renal cell carcinoma using interleukin-2 (aldesleukin) and interferon alfa. However, the immergence of new immunotherapies, such as anti-PD-1 and anti-CTLA-4 monoclonal antibodies, have expanded the role of immunotherapy to many other, more common, malignancies, such as lung cancer, breast cancer, prostate cancer, head and neck cancer, and many more.

Most randomized clinical trials study drugs as second- or occasionally third-line therapy. However, many patients continue to be treated beyond the third-line setting, often without evidence-based data to support potential benefit. Patients often place value on treatments unlikely to work so as not to give up hope. These “hopeful gambles,” even with the potential of significant toxicity and decreased quality of life (QOL), are common in cancer treatment.6 In addition, oncologists often overestimate the clinical benefit when considering additional therapy in this setting.7

Influx of New Patients

Outside the VHA setting, the financial burden of cancer treatment has led to an influx of new patients transferring care to the VHA to reduce out-of-pocket expenses. Because private insurance copays for oral agents are increasing, many reaching 20% to 30%, out-of-pocket expenses for medications can reach several thousand dollars per month. Patients often change insurance plans due to changing jobs or to decrease cost, or employers may change plans to save money, which may significantly alter or discontinue coverage. Patients often request that the VA provide medication while continuing to see only their private oncologist. This practice should be discouraged because the VA, without clinical involvement, may supply drugs for inappropriate indications. In addition, VA providers writing prescriptions for medications without personally following patients may be liable for poor outcomes.

VA PBM Services

Prior to 1995, the VA was a much criticized and poorly performing health care system that had experienced significant budget cuts, forcing many veterans to seek care outside the VA. Then beginning in 1995, a remarkable transformation occurred, which modernized and improved the VA into a system that consistently outperforms the private sector in quality of care, patient safety, patient satisfaction, all at a lower cost.8 The story of the VA’s transformation has been well chronicled by Phillip Longman.9

Under the direction of VA Under Secretary for Health Kenneth Kizer, MD, MPH, VA established PBM Clinical Services to develop and maintain the National Drug Formulary, create clinical guidance documents, and manage drug costs and utilization. A recent article by Heron and Geraci examined the functions and role of the VA PBM in controlling oncology drug costs.10 The following is a brief review of several documents and VA PBM responsibilities as reviewed by Heron and Geraci.

VA National Formulary

Prior to the establishment of the VA National Formulary in 1995, each VA maintained its own formulary, which led to extreme variability in drug access across the country. When a patient accessed care at different VAMCs, it was common for the patient’s medications to be changed based on the specific facility formulary. This practice led to many potential problems, such as lack of clinical benefit and potentially increased or new toxicities, and led to extra hospital visits for monitoring and adjustment of medications.

 

 

In contrast, the VA National Formulary now offers a uniform pharmacy benefit to all veterans by reducing variation in access to drugs. In addition, using preferred agents in each drug class provides VA with additional leverage when contracting with drug suppliers to reduce prices across the entire VA system.

Many oncology agents are not included on the VA National Formulary due to cost and the potential for off-label use. However, the formulary status of oncology agents in no way limits access or the availability of any oncology drug for appropriate patients. In fact, nonformulary approval requests work as a mechanism for review to ensure that these agents are used properly in the subset of patients who are most likely to benefit.

The PBM assesses all new oncology drugs for value and potential use within the VA, as well as cost impact. Following this assessment, various clinical guidance documents may be developed that are intended to guide clinicians in the proper use of medications for veterans. All documents prepared by the PBM undergo an extensive peer review by the Medical Advisory Panel and other experts in the field.

 

Drug Monographs

A drug monograph is a comprehensive, evidence-based drug review that summarizes efficacy and safety based on clinical trial data published in peer-reviewed journals, abstracts, and/or FDA Medical Review transcripts. Cost-effectiveness analysis is included if available.

Criteria for Use

Criteria for Use (CFU) are developed for drugs considered to be at high risk for inappropriate use or with safety concerns. The purpose of the CFU is to select patients most likely to benefit from these agents by using clinical criteria, which may qualify or eliminate a patient for treatment. National CFUs are available on the national PBM website. Local CFUs are often written and shared among oncology pharmacists via the VA oncology pharmacist listserv.

Abbreviated Reviews

Similar to drug monographs, abbreviated reviews are much shorter and focus on the relevant clinical sections of the drug monograph necessary for clinical or formulary decision making.

National Acquisition Center

The National Acquisition Center (NAC) is the pharmaceutical contracting mechanism for the VA and works closely with the PBM.5 The NAC pursues significant drug price reductions for the VA based on many strategies. Public Law 102-585 ensures that certain government agencies, including the VA, receive special discounts on pharmaceuticals, which is at least a 24% discount from the nonfederal Average Manufacturer Price. This is known as the Federal Supply Schedule (FSS) and/or Big 4 pricing. In addition, bulk purchases and performance-based incentive agreements can lead to substantial local discounts. By working with specific drug distribution and warehouse contractors, the NAC assures ready access to drugs for VA patients. The NAC also allows for an efficient drug inventory process, thus reducing inventory management costs.

Guidance Documents

In 2012, the VA Oncology Field Advisory Committee (FAC) created the High Cost Oncology Drug Work Group to address the impact of high-cost oncology drugs within the VA.11 This work group was composed of VA oncologists and pharmacists whose efforts resulted in 5 guidance documents designed to reduce drug costs by optimizing therapy and reducing waste: (1) Dose Rounding in Oncology; (2) Oral Anticancer Drugs Dispensing and Monitoring; (3) Oncology Drug Table: Recommended Dispensing and Monitoring; (4) Chemotherapy Review Committee Process; and (5) Determining Clinical Benefit of High Cost Oncology Drugs. Reviews of 2 of these documents follows.

Determining Clinical Benefit of High Cost Oncology Drugs provides a decision tool to aid members of the oncology health care team in optimizing patient outcomes while attempting to obtain the greatest value from innovative therapies. When a high-cost or off-label request is made for a particular patient, using this process encourages thoughtful and evidence-based use of the drug by considering all clinical evidence in addition to the FDA-approved indication. Finally, a drug’s safety profile in relation to the indication, therapeutic goal, and specific patient characteristics and desires are integrated into a final decision to determine the appropriateness of the therapeutic intervention for the patient.

Oncology Drug Table: Recommended Dispensing and Monitoring contains a list of oral oncology drugs and includes recommendations for dispensing amount, adverse effects, laboratory monitoring, formulary status, approval requirements, and monthly cost of each agent based on the current NAC pricing.5 Cost awareness is critical when comparing alternative treatment options to minimize cost when treatments with similar benefits are considered. Most VA oncologists do not have easy access to the cost of various treatments and can be surprised about how expensive many common regimens cost. The costs listed in this document are updated about every 3 months.

 

 

Conclusion

Using newer, expensive targeted oncology agents in a cost-effective manner must be a proactive, collaborative, and multidisciplinary process. Pharmacists should not be solely responsible for monitoring and controlling high-cost treatments. Well-informed, evidence-based decisions are needed to ensure expensive agents are used in the subset of patients who are most likely to benefit. Clinical tools addressing value should be used to aid in appropriate and cost-effective treatment plans using drug monographs and CFUs, VHA Guidance on Determining Clinical Benefit of High Cost Oncology Drugs, and the Oral Chemotherapy Dispensing and Monitoring Reference, among other resources. Due to the subjective nature of value in medicine, agreeing on policy will have many challenges, such as how to place a value on various gains in overall survival, progression free survival, response rates, and QOL.

eAppendix

The Cost of Oncology Drugs: A Pharmacy Perspective, Part 1, appeared in the Federal Practitioner February 2016 special issue “Best Practices in Hematology and Oncology” and can be accessed here.

Health care costs are the fastest growing financial segment of the U.S. economy. The cost of medications, especially those for treating cancer, is the leading cause of increased health care spending.1 Until recently, the discussion of the high costs of cancer treatment was rarely made public.

Part 1 of this article focused on the emerging discussion of the financial impact of high-cost drugs in the U.S. Part 2 will focus on the drivers of increasing oncology drug costs and the challenges high-cost medications pose for the VA. The article also will review the role of the VA Pharmacy Benefits Management Service (PBM) in evaluating new oncology agents. Also presented are the clinical guidance tools designed to aid the clinician in the cost-effective use of these agents and results of a nationwide survey of VA oncology pharmacists regarding the use of cost-containment strategies.

 

Cost Drivers

Many factors are driving increased oncology drug costs within the VA. Although the cost of individual drugs has the largest impact on the accelerating cost of treating each patient, other clinical and social factors may play a role.

Increasing Cost of Individual Drugs

Drug pricing is not announced until after FDA approval. Oncology drugs at the high end of the cost spectrum are rarely curative and often add only weeks or months to overall survival (OS), the gold standard. Current clinical trial design often uses progression free survival (PFS) as the primary endpoint, which makes the use of traditional pharmacoeconomic determinations of value difficult. In addition, many new drugs are first in class and/or have narrow indications that preclude competition from other drugs. Although addressing the issue of the market price for drugs seems to be one that is not controllable, there is increasing demand for drug pricing reform.2

Many believe drug prices should be linked directly to clinical benefit. In a recent article, Goldstein and colleagues proposed establishing a value-based price for necitumumab based on clinical benefit, prior to FDA approval.3 When this analysis was done, necitumumab was pending FDA approval in combination with cisplatin and gemcitabine for the treatment of squamous carcinoma of the lung. Using clinical data from the SQUIRE trial on which FDA approval was based, the addition of necitumumab to the chemotherapy regimen led to an incremental survival benefit of 0.15 life-years and 0.11 quality-adjusted life-years (QALY).4 Using a Markov model to evaluate cost-effectiveness, these authors established that the price of necitumumab should be from $563 to $1,309 per cycle. Necitumumab was approved by the FDA on November 24, 2015, with the VA acquisition cost, as of May 2016, at $6,100 per cycle.

Lack of Generic Products

The approval of generic alternatives for targeted oncology agents should reduce the cost of treating oncology patients. However, since imatinib was approved in May 2001, no single targeted agent had become available as a generic until February 1, 2016, when generic imatinib was made available in the U.S. following approval by the FDA. Currently, generic imatinib is not used in the VA due to lack of Federal Supply Schedule (FSS) contract pricing, which leads to a generic cost that is much higher than the brand-name drug, Gleevec ($6,127 per month vs $9,472 per month for the generic). The reality is that many older agents have steadily increased in price, outpacing inflation (Table 1).5

Aging U.S. Population

Advancing age is the most common risk factor for cancer, leading to an increase in the incidence and treatment of cancer. Because many newer agents are considered easier to tolerate than are traditional cytotoxic chemotherapy, clinicians have become more comfortable treating elderly patients, and geriatric oncology has become an established subspecialty within oncology.

Changing Treatment Paradigms

The use of targeted therapies is changing the paradigm from the acute treatment of cancer to chronic cancer management. Most targeted therapies are continued until disease progression or toxicity, leading to chronic, open-ended treatment. This approach is in contrast to older treatment approaches such as chemotherapy, which is often given for a limited duration followed by observation. When successful, chronic treatment with targeted agents can lead to unanticipated high costs. The following current cases at the VA San Diego Healthcare System illustrate this point:

  • Renal cell carcinoma: 68-year-old man diagnosed in 2005 with a recurrence in 2012

- High-dose interleukin-2 (2 cycles); sunitinib (3.3 years); pazopanib (2 months); everolimus (2 months); sorafenib (3 months); axitinib (7 months)

- Now contemplating anti-PD-1, pending emerging data. Total cost to date ~ $324,300.
 

 

  • Cutaneous T-cell lymphoma: 68-year-old man started romidepsin September 22, 2010
- Received 108 doses through May 2016. Total cost to date~ $652,970.

The rate of FDA approval for oncology drugs has been accelerating rapidly in the past 15 years. Sequential therapies beyond second-line therapy are common as more agents become available. Table 2 shows FDA approval for all cancer drugs by decade.

As researchers continue to better understand the many pathways involved with the development and progression of cancer, they are beginning to combine multiple targeted agents to augment response rates, prolong survival, and reduce the potential for resistance. Recent combination regimens approved by the FDA include dabrafenib plus trametinib (January 2014), and ipilimumab plus nivolumab (October 2015), both for the treatment of melanoma. In November 2015, ixazomib was FDA approved to be used in combination with lenalidomide for multiple myeloma. Many more combination regimens are currently in clinical trials, and more combinations are expected to receive FDA approval. It is easy to see how the combination of multiple expensive agents with the prospect of prolonged therapy has the potential to increase the cost of many regimens to well over $100,000 per year.

Maintenance therapy is used to prolong PFS for patients receiving an excellent response to primary therapy. For example, VA costs for maintenance regimens include lenalidomide 10 mg daily: $8,314 for 28 days equals $216,177 for 2 years; bortezomib 1.3 mg/m2 (2.6 mg) q: 2 weeks equals $60,730 for 2 years (includes waste as bortezomib 3.5-mg vials do not a contain preservative and must be discarded within 8 hours of preparation); and rituximab 800 mg q: 2 months equals $47,635 for 2 years.

Until recently, immunotherapy for cancer was limited to melanoma and renal cell carcinoma using interleukin-2 (aldesleukin) and interferon alfa. However, the immergence of new immunotherapies, such as anti-PD-1 and anti-CTLA-4 monoclonal antibodies, have expanded the role of immunotherapy to many other, more common, malignancies, such as lung cancer, breast cancer, prostate cancer, head and neck cancer, and many more.

Most randomized clinical trials study drugs as second- or occasionally third-line therapy. However, many patients continue to be treated beyond the third-line setting, often without evidence-based data to support potential benefit. Patients often place value on treatments unlikely to work so as not to give up hope. These “hopeful gambles,” even with the potential of significant toxicity and decreased quality of life (QOL), are common in cancer treatment.6 In addition, oncologists often overestimate the clinical benefit when considering additional therapy in this setting.7

Influx of New Patients

Outside the VHA setting, the financial burden of cancer treatment has led to an influx of new patients transferring care to the VHA to reduce out-of-pocket expenses. Because private insurance copays for oral agents are increasing, many reaching 20% to 30%, out-of-pocket expenses for medications can reach several thousand dollars per month. Patients often change insurance plans due to changing jobs or to decrease cost, or employers may change plans to save money, which may significantly alter or discontinue coverage. Patients often request that the VA provide medication while continuing to see only their private oncologist. This practice should be discouraged because the VA, without clinical involvement, may supply drugs for inappropriate indications. In addition, VA providers writing prescriptions for medications without personally following patients may be liable for poor outcomes.

VA PBM Services

Prior to 1995, the VA was a much criticized and poorly performing health care system that had experienced significant budget cuts, forcing many veterans to seek care outside the VA. Then beginning in 1995, a remarkable transformation occurred, which modernized and improved the VA into a system that consistently outperforms the private sector in quality of care, patient safety, patient satisfaction, all at a lower cost.8 The story of the VA’s transformation has been well chronicled by Phillip Longman.9

Under the direction of VA Under Secretary for Health Kenneth Kizer, MD, MPH, VA established PBM Clinical Services to develop and maintain the National Drug Formulary, create clinical guidance documents, and manage drug costs and utilization. A recent article by Heron and Geraci examined the functions and role of the VA PBM in controlling oncology drug costs.10 The following is a brief review of several documents and VA PBM responsibilities as reviewed by Heron and Geraci.

VA National Formulary

Prior to the establishment of the VA National Formulary in 1995, each VA maintained its own formulary, which led to extreme variability in drug access across the country. When a patient accessed care at different VAMCs, it was common for the patient’s medications to be changed based on the specific facility formulary. This practice led to many potential problems, such as lack of clinical benefit and potentially increased or new toxicities, and led to extra hospital visits for monitoring and adjustment of medications.

 

 

In contrast, the VA National Formulary now offers a uniform pharmacy benefit to all veterans by reducing variation in access to drugs. In addition, using preferred agents in each drug class provides VA with additional leverage when contracting with drug suppliers to reduce prices across the entire VA system.

Many oncology agents are not included on the VA National Formulary due to cost and the potential for off-label use. However, the formulary status of oncology agents in no way limits access or the availability of any oncology drug for appropriate patients. In fact, nonformulary approval requests work as a mechanism for review to ensure that these agents are used properly in the subset of patients who are most likely to benefit.

The PBM assesses all new oncology drugs for value and potential use within the VA, as well as cost impact. Following this assessment, various clinical guidance documents may be developed that are intended to guide clinicians in the proper use of medications for veterans. All documents prepared by the PBM undergo an extensive peer review by the Medical Advisory Panel and other experts in the field.

 

Drug Monographs

A drug monograph is a comprehensive, evidence-based drug review that summarizes efficacy and safety based on clinical trial data published in peer-reviewed journals, abstracts, and/or FDA Medical Review transcripts. Cost-effectiveness analysis is included if available.

Criteria for Use

Criteria for Use (CFU) are developed for drugs considered to be at high risk for inappropriate use or with safety concerns. The purpose of the CFU is to select patients most likely to benefit from these agents by using clinical criteria, which may qualify or eliminate a patient for treatment. National CFUs are available on the national PBM website. Local CFUs are often written and shared among oncology pharmacists via the VA oncology pharmacist listserv.

Abbreviated Reviews

Similar to drug monographs, abbreviated reviews are much shorter and focus on the relevant clinical sections of the drug monograph necessary for clinical or formulary decision making.

National Acquisition Center

The National Acquisition Center (NAC) is the pharmaceutical contracting mechanism for the VA and works closely with the PBM.5 The NAC pursues significant drug price reductions for the VA based on many strategies. Public Law 102-585 ensures that certain government agencies, including the VA, receive special discounts on pharmaceuticals, which is at least a 24% discount from the nonfederal Average Manufacturer Price. This is known as the Federal Supply Schedule (FSS) and/or Big 4 pricing. In addition, bulk purchases and performance-based incentive agreements can lead to substantial local discounts. By working with specific drug distribution and warehouse contractors, the NAC assures ready access to drugs for VA patients. The NAC also allows for an efficient drug inventory process, thus reducing inventory management costs.

Guidance Documents

In 2012, the VA Oncology Field Advisory Committee (FAC) created the High Cost Oncology Drug Work Group to address the impact of high-cost oncology drugs within the VA.11 This work group was composed of VA oncologists and pharmacists whose efforts resulted in 5 guidance documents designed to reduce drug costs by optimizing therapy and reducing waste: (1) Dose Rounding in Oncology; (2) Oral Anticancer Drugs Dispensing and Monitoring; (3) Oncology Drug Table: Recommended Dispensing and Monitoring; (4) Chemotherapy Review Committee Process; and (5) Determining Clinical Benefit of High Cost Oncology Drugs. Reviews of 2 of these documents follows.

Determining Clinical Benefit of High Cost Oncology Drugs provides a decision tool to aid members of the oncology health care team in optimizing patient outcomes while attempting to obtain the greatest value from innovative therapies. When a high-cost or off-label request is made for a particular patient, using this process encourages thoughtful and evidence-based use of the drug by considering all clinical evidence in addition to the FDA-approved indication. Finally, a drug’s safety profile in relation to the indication, therapeutic goal, and specific patient characteristics and desires are integrated into a final decision to determine the appropriateness of the therapeutic intervention for the patient.

Oncology Drug Table: Recommended Dispensing and Monitoring contains a list of oral oncology drugs and includes recommendations for dispensing amount, adverse effects, laboratory monitoring, formulary status, approval requirements, and monthly cost of each agent based on the current NAC pricing.5 Cost awareness is critical when comparing alternative treatment options to minimize cost when treatments with similar benefits are considered. Most VA oncologists do not have easy access to the cost of various treatments and can be surprised about how expensive many common regimens cost. The costs listed in this document are updated about every 3 months.

 

 

Conclusion

Using newer, expensive targeted oncology agents in a cost-effective manner must be a proactive, collaborative, and multidisciplinary process. Pharmacists should not be solely responsible for monitoring and controlling high-cost treatments. Well-informed, evidence-based decisions are needed to ensure expensive agents are used in the subset of patients who are most likely to benefit. Clinical tools addressing value should be used to aid in appropriate and cost-effective treatment plans using drug monographs and CFUs, VHA Guidance on Determining Clinical Benefit of High Cost Oncology Drugs, and the Oral Chemotherapy Dispensing and Monitoring Reference, among other resources. Due to the subjective nature of value in medicine, agreeing on policy will have many challenges, such as how to place a value on various gains in overall survival, progression free survival, response rates, and QOL.

eAppendix

References

1. Bach PB. Limits on Medicare's ability to control rising spending on cancer drugs. N Engl J Med. 2009;360(6):626-633.

2. Kantarjian H, Steensma D, Rius Sanjuan J, Eishaug A, Light D. High cancer drug prices in the United States: reasons and proposed solutions. J Oncol Pract. 2014;10(4):e208-e211.

3. Goldstein DA, Chen Q, Ayer T, et al. Necitumumab in metastatic squamous cell lung cancer: establishing a value-based cost. JAMA Oncol. 2015;1(9):1293-1300.

4. Thatcher N, Hirsch FR, Luft AV, et al; SQUIRE Investigators. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015;16(7):763-774.

5. U.S. Department of Veterans Affairs, National Acquisition Center, Pharmaceutical Catalog Search. U.S. Department of Veterans Affairs, National Acquisition Center website. http://www1.va.gov/nac/index.cfm?template=Search_Pharmaceutical_Catalog. Updated June 13, 2016. Accessed June 13, 2016.

6. Lakdawalla DN, Romley JA, Sanchez Y, Maclean JR, Penrod JR, Philipson T. How cancer patients value hope and the implications for cost-effectiveness assessments of high-cost cancer therapies. Health Aff (Millwood). 2012;31(4):676-682.

7. Ubel PA, Berry SR, Nadler E, et al. In a survey, marked inconsistency in how oncologists judged value of high-cost cancer drugs in relation to gains in survival. Health Aff (Millwood). 2012;31(4):709-717.

8. Asch SM, McGlynn EA, Hogan MM, et al. Comparison of quality of care for patients in the Veterans Health Administration and patients in a national sample. Ann Intern Med. 2004;141(12):938-945. 9. Longman P. Best Care Anywhere: Why VA Health Care Would Work for Everyone. 3rd ed. San Francisco, CA: Berrett-Koehler Publishers; 2012. 10.  Heron BB, Geraci MC. Controlling the cost of oncology drugs within the VA: a national perspective. Fed Pract. 2015;32(suppl 1):18S-22S.

11.  U.S. Department of Veterans Affairs. Pharmacy Benefits Management Services Intranet, Documents and Lists. https://vaww.cmopnational.va.gov/cmop/PBM/Clinical%20Guidance/Forms/AllItems.aspx. Accessed May 19, 2016.

References

1. Bach PB. Limits on Medicare's ability to control rising spending on cancer drugs. N Engl J Med. 2009;360(6):626-633.

2. Kantarjian H, Steensma D, Rius Sanjuan J, Eishaug A, Light D. High cancer drug prices in the United States: reasons and proposed solutions. J Oncol Pract. 2014;10(4):e208-e211.

3. Goldstein DA, Chen Q, Ayer T, et al. Necitumumab in metastatic squamous cell lung cancer: establishing a value-based cost. JAMA Oncol. 2015;1(9):1293-1300.

4. Thatcher N, Hirsch FR, Luft AV, et al; SQUIRE Investigators. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015;16(7):763-774.

5. U.S. Department of Veterans Affairs, National Acquisition Center, Pharmaceutical Catalog Search. U.S. Department of Veterans Affairs, National Acquisition Center website. http://www1.va.gov/nac/index.cfm?template=Search_Pharmaceutical_Catalog. Updated June 13, 2016. Accessed June 13, 2016.

6. Lakdawalla DN, Romley JA, Sanchez Y, Maclean JR, Penrod JR, Philipson T. How cancer patients value hope and the implications for cost-effectiveness assessments of high-cost cancer therapies. Health Aff (Millwood). 2012;31(4):676-682.

7. Ubel PA, Berry SR, Nadler E, et al. In a survey, marked inconsistency in how oncologists judged value of high-cost cancer drugs in relation to gains in survival. Health Aff (Millwood). 2012;31(4):709-717.

8. Asch SM, McGlynn EA, Hogan MM, et al. Comparison of quality of care for patients in the Veterans Health Administration and patients in a national sample. Ann Intern Med. 2004;141(12):938-945. 9. Longman P. Best Care Anywhere: Why VA Health Care Would Work for Everyone. 3rd ed. San Francisco, CA: Berrett-Koehler Publishers; 2012. 10.  Heron BB, Geraci MC. Controlling the cost of oncology drugs within the VA: a national perspective. Fed Pract. 2015;32(suppl 1):18S-22S.

11.  U.S. Department of Veterans Affairs. Pharmacy Benefits Management Services Intranet, Documents and Lists. https://vaww.cmopnational.va.gov/cmop/PBM/Clinical%20Guidance/Forms/AllItems.aspx. Accessed May 19, 2016.

Issue
Federal Practitioner - 33(7)
Issue
Federal Practitioner - 33(7)
Page Number
35-39
Page Number
35-39
Publications
Publications
Topics
Article Type
Display Headline
The Cost of Oncology Drugs: A Pharmacy Perspective, Part 2
Display Headline
The Cost of Oncology Drugs: A Pharmacy Perspective, Part 2
Legacy Keywords
oncology, medication, VA National Formulary, cancer treatment, imatinib
Legacy Keywords
oncology, medication, VA National Formulary, cancer treatment, imatinib
Sections
Disallow All Ads
Alternative CME
Use ProPublica
Article PDF Media

SU2C announces researcher-industry collaboration on immunotherapy

Article Type
Changed
Fri, 01/04/2019 - 09:52
Display Headline
SU2C announces researcher-industry collaboration on immunotherapy

Stand Up To Cancer is calling for proposals to investigate additional uses for nivolumab, ipilimumab, elotuzumab, and urelumab, as part of a new researcher-industry collaborative program.

As many as four projects will be funded by Bristol-Myers Squibb, maker of the four agents, in the range of $1 million to $3 million each, according to a written statement from the American Association for Cancer Research (AACR).

The company will provide access to the three drugs already approved for the treatement of various cancers –nivolumab, ipilimumab, and elotuzumab– and to urelumab, an investigational agent that is currently in early clinical trials.

Proposals can include the study of one or more of the products, alone or in combination with other treatments, and may include products from other companies, as well as explore potential new uses for the drug(s), AACR said in the statement.

Nivolumab (Opdivo) is currently approved to treat advanced melanoma, non-small cell lung cancer, renal cell carcinoma, and classical Hodgkin lymphoma; Ipilimumab (Yervoy) is approved to treat melanoma; and elotuzumab (Empliciti) is approved to treat multiple myeloma, in conjunction with other drugs. Urelumab is being evaluated as a treatment for a range of cancers, including some hematological cancers, advanced colorectal cancer, and head and neck cancers.

The Stand Up To Cancer (SU2C) Catalyst program was launched in April to “use funding and materials from the pharmaceutical, biotechnology, diagnostic, and medical devices industries to accelerate research on cancer prevention, detection, and treatment,” according to a written statement from SU2C. Founding collaborators in addition to Bristol-Myers Squibb include Merck and Genentech.

The Catalyst projects must follow the SU2C model be carried out by a collaborative team, and be designed to accelerate the clinical use of therapeutic agents within the 3-year term of the grant, and to deliver near-term patient benefit.

The Request for Proposal for the Bristol-Myers Squibb agents is available at proposalCENTRAL, with proposals due by noon ET Monday, Aug. 15.

[email protected]

On Twitter @NikolaidesLaura

Publications
Topics

Stand Up To Cancer is calling for proposals to investigate additional uses for nivolumab, ipilimumab, elotuzumab, and urelumab, as part of a new researcher-industry collaborative program.

As many as four projects will be funded by Bristol-Myers Squibb, maker of the four agents, in the range of $1 million to $3 million each, according to a written statement from the American Association for Cancer Research (AACR).

The company will provide access to the three drugs already approved for the treatement of various cancers –nivolumab, ipilimumab, and elotuzumab– and to urelumab, an investigational agent that is currently in early clinical trials.

Proposals can include the study of one or more of the products, alone or in combination with other treatments, and may include products from other companies, as well as explore potential new uses for the drug(s), AACR said in the statement.

Nivolumab (Opdivo) is currently approved to treat advanced melanoma, non-small cell lung cancer, renal cell carcinoma, and classical Hodgkin lymphoma; Ipilimumab (Yervoy) is approved to treat melanoma; and elotuzumab (Empliciti) is approved to treat multiple myeloma, in conjunction with other drugs. Urelumab is being evaluated as a treatment for a range of cancers, including some hematological cancers, advanced colorectal cancer, and head and neck cancers.

The Stand Up To Cancer (SU2C) Catalyst program was launched in April to “use funding and materials from the pharmaceutical, biotechnology, diagnostic, and medical devices industries to accelerate research on cancer prevention, detection, and treatment,” according to a written statement from SU2C. Founding collaborators in addition to Bristol-Myers Squibb include Merck and Genentech.

The Catalyst projects must follow the SU2C model be carried out by a collaborative team, and be designed to accelerate the clinical use of therapeutic agents within the 3-year term of the grant, and to deliver near-term patient benefit.

The Request for Proposal for the Bristol-Myers Squibb agents is available at proposalCENTRAL, with proposals due by noon ET Monday, Aug. 15.

[email protected]

On Twitter @NikolaidesLaura

Stand Up To Cancer is calling for proposals to investigate additional uses for nivolumab, ipilimumab, elotuzumab, and urelumab, as part of a new researcher-industry collaborative program.

As many as four projects will be funded by Bristol-Myers Squibb, maker of the four agents, in the range of $1 million to $3 million each, according to a written statement from the American Association for Cancer Research (AACR).

The company will provide access to the three drugs already approved for the treatement of various cancers –nivolumab, ipilimumab, and elotuzumab– and to urelumab, an investigational agent that is currently in early clinical trials.

Proposals can include the study of one or more of the products, alone or in combination with other treatments, and may include products from other companies, as well as explore potential new uses for the drug(s), AACR said in the statement.

Nivolumab (Opdivo) is currently approved to treat advanced melanoma, non-small cell lung cancer, renal cell carcinoma, and classical Hodgkin lymphoma; Ipilimumab (Yervoy) is approved to treat melanoma; and elotuzumab (Empliciti) is approved to treat multiple myeloma, in conjunction with other drugs. Urelumab is being evaluated as a treatment for a range of cancers, including some hematological cancers, advanced colorectal cancer, and head and neck cancers.

The Stand Up To Cancer (SU2C) Catalyst program was launched in April to “use funding and materials from the pharmaceutical, biotechnology, diagnostic, and medical devices industries to accelerate research on cancer prevention, detection, and treatment,” according to a written statement from SU2C. Founding collaborators in addition to Bristol-Myers Squibb include Merck and Genentech.

The Catalyst projects must follow the SU2C model be carried out by a collaborative team, and be designed to accelerate the clinical use of therapeutic agents within the 3-year term of the grant, and to deliver near-term patient benefit.

The Request for Proposal for the Bristol-Myers Squibb agents is available at proposalCENTRAL, with proposals due by noon ET Monday, Aug. 15.

[email protected]

On Twitter @NikolaidesLaura

Publications
Publications
Topics
Article Type
Display Headline
SU2C announces researcher-industry collaboration on immunotherapy
Display Headline
SU2C announces researcher-industry collaboration on immunotherapy
Disallow All Ads

Immunosuppressive regimens did not affect risk of cancer recurrence in meta-analysis

Consult an oncologist before using immunosuppressants in at-risk IBD patients
Article Type
Changed
Wed, 05/26/2021 - 13:54
Display Headline
Immunosuppressive regimens did not affect risk of cancer recurrence in meta-analysis

Among patients with immune-mediated diseases and a history of malignancy, cancer recurrence rates were similar regardless of whether they received tumor necrosis factor inhibitors, traditional immunosuppressants, or no immunosuppression, according to a meta-analysis of 16 cohort and case-control studies.

“However, there is a need for larger studies to prospectively monitor for cancer recurrence and new malignancies in this population to better inform our practice,” wrote Dr. Edward Shelton of Massachusetts General Hospital in Boston, and his associates. The report is in the July issue of Gastroenterology.

Patients with immune-mediated diseases often have a history of malignancy, raising questions about the effects of therapies that target the immune system, the researchers noted. However, relevant studies have been “small with few events, preventing robust estimates of risk,” they said. They searched Medline, Embase, and conference proceedings through April 2015 for studies of the risk of primary or recurrent cancer among patients with a history of malignancy who were exposed to thiopurines, methotrexate, or anti-TNF agents, or no immunosuppression. Among the 16 studies meeting these criteria, seven included patients with rheumatoid arthritis, seven included patients with inflammatory bowel disease, one included both diseases, and one included patients with psoriasis. Twelve were cohort studies, one was a case-control study, and three were case series. The resulting meta-analysis comprised 11,702 patients who contributed a total of 31,258 person-years of follow-up (Gastroenterology 2016 May 13. doi: 10.1053/j.gastro.2016.03.037).

Rates of cancer recurrence were statistically similar among patients who received no immunosuppression, anti-TNF therapy, traditional immune modulators, or combination regimens (P greater than .1 for differences among groups). Numerically, however, the risk of recurrent cancer was highest with combination immunotherapy (54.5 cases per 1,000 person-years of follow-up), compared with 37.5 cases per 1,000 person-years for patients who did not receive immunosuppressive treatment, 36.2 cases per 1,000 person-years for patients who received traditional immunomodulator monotherapy, and 33.8 cases per 1,000 person-years for patients who received anti-TNF agents. Analyses of subgroups of patients with new or recurrent cancers, or various types of therapies, and of specific immune-mediated diseases yielded “similar results, with no increase in risk,” said the researchers. Furthermore, rates of cancer recurrence did not statistically differ depending on whether patients started immunosuppression less than or more than 6 years after their index cancer (P = .43).

“Treatment decisions after a cancer diagnosis are complex, and must take into account the natural history of cancer, histologic type and stage, time from diagnosis, and course of underlying chronic inflammatory disease,” the researchers noted. “Our findings suggest that anti-TNF therapy, conventional immunosuppressant therapy, or combination immunosuppression are not associated with an increased risk of cancer recurrence in this population.”

The researchers cited several limitations. Notably, three of the studies included only 20 patients, and the studies included variable index cancers and methods for detecting recurrent cancers. “It also is possible, and likely, that patients who were at high risk for recurrence were not recommenced on immunosuppression, and consequently our findings may be more applicable to a population in which the patient and physicians were comfortable with re-initiation of therapy,” they said.

The National Institutes of Health supported several of the study investigators. Dr. Shelton had no disclosures. Five coinvestigators disclosed relationships with a number of pharmaceutical companies.

Click for Credit Link
Body

As the proportion of inflammatory bowel disease (IBD) patients who are elderly rises, so does the occurrence of coexisting cancers accompanying advanced age. Experience from the posttransplant population has prompted concerns that use of immunosuppression may heighten the risk of new or recurrent cancers in individuals who have a prior history of cancer.

 

Dr. Geoffrey Nguyen

This apprehension may partly explain why elderly IBD patients are much less likely to be treated with immunosuppressive therapies than their younger counterparts are. Prior studies in IBD populations have not shown any association between use of immunosuppressants and recurrent cancer in individuals with prior cancer. However, these studies may have been underpowered to detect such associations.

Dr. Shelton and his colleagues performed a meta-analysis that draws upon more than 16 studies of patients with immune-mediated diseases with prior history of cancer. The investigators confirmed no association between immunosuppression and recurrent or new cancers. Despite some methodologic limitations, the study’s findings support a body of literature that suggests no increased risk of recurrent malignancy with immunosuppression in immune-mediated diseases, including IBD.

The findings also provide additional support for recent ECCO clinical guidelines, which recommend that immunosuppressive therapy can be initiated in patients with cancer after a 2- to 5-year cancer-free interval, depending on the risk of recurrence associated with specific types of cancers. A key point is that the decision to use immunosuppressants in IBD patients with prior cancer should be made with input from an oncologist and be individualized by taking into account disease course, surgical alternatives, and risk of cancer recurrence.

Dr. Geoffrey Nguyen, Mount Sinai Centre for Inflammatory Bowel Disease, University of Toronto. He has no conflicts of interest.

Publications
Topics
Sections
Click for Credit Link
Click for Credit Link
Body

As the proportion of inflammatory bowel disease (IBD) patients who are elderly rises, so does the occurrence of coexisting cancers accompanying advanced age. Experience from the posttransplant population has prompted concerns that use of immunosuppression may heighten the risk of new or recurrent cancers in individuals who have a prior history of cancer.

 

Dr. Geoffrey Nguyen

This apprehension may partly explain why elderly IBD patients are much less likely to be treated with immunosuppressive therapies than their younger counterparts are. Prior studies in IBD populations have not shown any association between use of immunosuppressants and recurrent cancer in individuals with prior cancer. However, these studies may have been underpowered to detect such associations.

Dr. Shelton and his colleagues performed a meta-analysis that draws upon more than 16 studies of patients with immune-mediated diseases with prior history of cancer. The investigators confirmed no association between immunosuppression and recurrent or new cancers. Despite some methodologic limitations, the study’s findings support a body of literature that suggests no increased risk of recurrent malignancy with immunosuppression in immune-mediated diseases, including IBD.

The findings also provide additional support for recent ECCO clinical guidelines, which recommend that immunosuppressive therapy can be initiated in patients with cancer after a 2- to 5-year cancer-free interval, depending on the risk of recurrence associated with specific types of cancers. A key point is that the decision to use immunosuppressants in IBD patients with prior cancer should be made with input from an oncologist and be individualized by taking into account disease course, surgical alternatives, and risk of cancer recurrence.

Dr. Geoffrey Nguyen, Mount Sinai Centre for Inflammatory Bowel Disease, University of Toronto. He has no conflicts of interest.

Body

As the proportion of inflammatory bowel disease (IBD) patients who are elderly rises, so does the occurrence of coexisting cancers accompanying advanced age. Experience from the posttransplant population has prompted concerns that use of immunosuppression may heighten the risk of new or recurrent cancers in individuals who have a prior history of cancer.

 

Dr. Geoffrey Nguyen

This apprehension may partly explain why elderly IBD patients are much less likely to be treated with immunosuppressive therapies than their younger counterparts are. Prior studies in IBD populations have not shown any association between use of immunosuppressants and recurrent cancer in individuals with prior cancer. However, these studies may have been underpowered to detect such associations.

Dr. Shelton and his colleagues performed a meta-analysis that draws upon more than 16 studies of patients with immune-mediated diseases with prior history of cancer. The investigators confirmed no association between immunosuppression and recurrent or new cancers. Despite some methodologic limitations, the study’s findings support a body of literature that suggests no increased risk of recurrent malignancy with immunosuppression in immune-mediated diseases, including IBD.

The findings also provide additional support for recent ECCO clinical guidelines, which recommend that immunosuppressive therapy can be initiated in patients with cancer after a 2- to 5-year cancer-free interval, depending on the risk of recurrence associated with specific types of cancers. A key point is that the decision to use immunosuppressants in IBD patients with prior cancer should be made with input from an oncologist and be individualized by taking into account disease course, surgical alternatives, and risk of cancer recurrence.

Dr. Geoffrey Nguyen, Mount Sinai Centre for Inflammatory Bowel Disease, University of Toronto. He has no conflicts of interest.

Title
Consult an oncologist before using immunosuppressants in at-risk IBD patients
Consult an oncologist before using immunosuppressants in at-risk IBD patients

Among patients with immune-mediated diseases and a history of malignancy, cancer recurrence rates were similar regardless of whether they received tumor necrosis factor inhibitors, traditional immunosuppressants, or no immunosuppression, according to a meta-analysis of 16 cohort and case-control studies.

“However, there is a need for larger studies to prospectively monitor for cancer recurrence and new malignancies in this population to better inform our practice,” wrote Dr. Edward Shelton of Massachusetts General Hospital in Boston, and his associates. The report is in the July issue of Gastroenterology.

Patients with immune-mediated diseases often have a history of malignancy, raising questions about the effects of therapies that target the immune system, the researchers noted. However, relevant studies have been “small with few events, preventing robust estimates of risk,” they said. They searched Medline, Embase, and conference proceedings through April 2015 for studies of the risk of primary or recurrent cancer among patients with a history of malignancy who were exposed to thiopurines, methotrexate, or anti-TNF agents, or no immunosuppression. Among the 16 studies meeting these criteria, seven included patients with rheumatoid arthritis, seven included patients with inflammatory bowel disease, one included both diseases, and one included patients with psoriasis. Twelve were cohort studies, one was a case-control study, and three were case series. The resulting meta-analysis comprised 11,702 patients who contributed a total of 31,258 person-years of follow-up (Gastroenterology 2016 May 13. doi: 10.1053/j.gastro.2016.03.037).

Rates of cancer recurrence were statistically similar among patients who received no immunosuppression, anti-TNF therapy, traditional immune modulators, or combination regimens (P greater than .1 for differences among groups). Numerically, however, the risk of recurrent cancer was highest with combination immunotherapy (54.5 cases per 1,000 person-years of follow-up), compared with 37.5 cases per 1,000 person-years for patients who did not receive immunosuppressive treatment, 36.2 cases per 1,000 person-years for patients who received traditional immunomodulator monotherapy, and 33.8 cases per 1,000 person-years for patients who received anti-TNF agents. Analyses of subgroups of patients with new or recurrent cancers, or various types of therapies, and of specific immune-mediated diseases yielded “similar results, with no increase in risk,” said the researchers. Furthermore, rates of cancer recurrence did not statistically differ depending on whether patients started immunosuppression less than or more than 6 years after their index cancer (P = .43).

“Treatment decisions after a cancer diagnosis are complex, and must take into account the natural history of cancer, histologic type and stage, time from diagnosis, and course of underlying chronic inflammatory disease,” the researchers noted. “Our findings suggest that anti-TNF therapy, conventional immunosuppressant therapy, or combination immunosuppression are not associated with an increased risk of cancer recurrence in this population.”

The researchers cited several limitations. Notably, three of the studies included only 20 patients, and the studies included variable index cancers and methods for detecting recurrent cancers. “It also is possible, and likely, that patients who were at high risk for recurrence were not recommenced on immunosuppression, and consequently our findings may be more applicable to a population in which the patient and physicians were comfortable with re-initiation of therapy,” they said.

The National Institutes of Health supported several of the study investigators. Dr. Shelton had no disclosures. Five coinvestigators disclosed relationships with a number of pharmaceutical companies.

Among patients with immune-mediated diseases and a history of malignancy, cancer recurrence rates were similar regardless of whether they received tumor necrosis factor inhibitors, traditional immunosuppressants, or no immunosuppression, according to a meta-analysis of 16 cohort and case-control studies.

“However, there is a need for larger studies to prospectively monitor for cancer recurrence and new malignancies in this population to better inform our practice,” wrote Dr. Edward Shelton of Massachusetts General Hospital in Boston, and his associates. The report is in the July issue of Gastroenterology.

Patients with immune-mediated diseases often have a history of malignancy, raising questions about the effects of therapies that target the immune system, the researchers noted. However, relevant studies have been “small with few events, preventing robust estimates of risk,” they said. They searched Medline, Embase, and conference proceedings through April 2015 for studies of the risk of primary or recurrent cancer among patients with a history of malignancy who were exposed to thiopurines, methotrexate, or anti-TNF agents, or no immunosuppression. Among the 16 studies meeting these criteria, seven included patients with rheumatoid arthritis, seven included patients with inflammatory bowel disease, one included both diseases, and one included patients with psoriasis. Twelve were cohort studies, one was a case-control study, and three were case series. The resulting meta-analysis comprised 11,702 patients who contributed a total of 31,258 person-years of follow-up (Gastroenterology 2016 May 13. doi: 10.1053/j.gastro.2016.03.037).

Rates of cancer recurrence were statistically similar among patients who received no immunosuppression, anti-TNF therapy, traditional immune modulators, or combination regimens (P greater than .1 for differences among groups). Numerically, however, the risk of recurrent cancer was highest with combination immunotherapy (54.5 cases per 1,000 person-years of follow-up), compared with 37.5 cases per 1,000 person-years for patients who did not receive immunosuppressive treatment, 36.2 cases per 1,000 person-years for patients who received traditional immunomodulator monotherapy, and 33.8 cases per 1,000 person-years for patients who received anti-TNF agents. Analyses of subgroups of patients with new or recurrent cancers, or various types of therapies, and of specific immune-mediated diseases yielded “similar results, with no increase in risk,” said the researchers. Furthermore, rates of cancer recurrence did not statistically differ depending on whether patients started immunosuppression less than or more than 6 years after their index cancer (P = .43).

“Treatment decisions after a cancer diagnosis are complex, and must take into account the natural history of cancer, histologic type and stage, time from diagnosis, and course of underlying chronic inflammatory disease,” the researchers noted. “Our findings suggest that anti-TNF therapy, conventional immunosuppressant therapy, or combination immunosuppression are not associated with an increased risk of cancer recurrence in this population.”

The researchers cited several limitations. Notably, three of the studies included only 20 patients, and the studies included variable index cancers and methods for detecting recurrent cancers. “It also is possible, and likely, that patients who were at high risk for recurrence were not recommenced on immunosuppression, and consequently our findings may be more applicable to a population in which the patient and physicians were comfortable with re-initiation of therapy,” they said.

The National Institutes of Health supported several of the study investigators. Dr. Shelton had no disclosures. Five coinvestigators disclosed relationships with a number of pharmaceutical companies.

Publications
Publications
Topics
Article Type
Display Headline
Immunosuppressive regimens did not affect risk of cancer recurrence in meta-analysis
Display Headline
Immunosuppressive regimens did not affect risk of cancer recurrence in meta-analysis
Click for Credit Status
Active
Sections
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Vitals

Key clinical point: Among patients with immune-mediated diseases and a history of cancer, rates of cancer recurrence did not statistically differ according to whether or not they received immunosuppression regimens, or regimen type.

Major finding: Numerically, the rate was highest with combination immunotherapy (54.5 cases per 1,000 person-years of follow-up), vs. 37.5 cases per 1,000 person-years for no immunosuppression, 36.2 cases per 1,000 person-years for traditional immunomodulator monotherapy, and 33.8 cases per 1,000 person-years for anti-TNF agents.

Data source: A systematic review and meta-analysis of 16 studies and 11,702 patients with rheumatoid arthritis, inflammatory bowel disease, and psoriasis.

Disclosures: The National Institutes of Health supported several of the study investigators. Dr. Shelton had no disclosures. Five coinvestigators disclosed relationships with a number of pharmaceutical companies.

Pembrolizumab paired with immunostimulator is safe and tolerable

Article Type
Changed
Fri, 01/04/2019 - 13:17
Display Headline
Pembrolizumab paired with immunostimulator is safe and tolerable

CHICAGO – Combining an immunostimulatory agent with the PD-1 checkpoint inhibitor pembrolizumab appeared quite safe and very tolerable, in a small phase Ib study.

There were some signs of efficacy against a variety of solid tumors, as well as biomarker trends showing immune activity.

In the phase Ib trial, researchers combined escalating doses (0.45-5.0 mg/kg) of PF-2566, an investigative immunostimulatory agent, with the anti–PD-1 checkpoint inhibitor pembrolizumab at 2 mg/kg, with both drugs given intravenously once every 3 weeks for a maximum of 32 cycles. A primary objective of the trial was to determine a maximum tolerated dose. Secondary objectives were to assess safety and tolerability and to determine any antitumor responses.

 

Dr. Anthony Tolcher

PF-2566 (Utomilumab/PF-05082566) is a monoclonal agonist targeting 4-1BB, a “costimulatory molecule that’s induced upon T-cell receptor activation and ultimately enhances cytotoxic T-cell response and effector status,” said Dr. Anthony Tolcher of the START Center for Cancer Care, San Antonio, at the annual meeting of the American Society of Clinical Oncology.

Eligible patients were 18 years or older, had a performance status of 0-1, and had advanced or metastatic solid tumors that had progressed on standard therapy or for which no standard therapy was available. They could not have had any form of immunosuppressive therapy in the 2 weeks prior to registration, a monoclonal antibody in the 2 months before the first dose, or any symptomatic or progressing central nervous system primary malignancies. Prior pembrolizumab was permitted.

Twenty-three patients (14 males) were heavily pretreated with a median of three prior therapies (range 0-9) for a variety of cancers, including six non–small-cell lung, five renal cell, three head and neck, and two each pancreatic and thyroid cancers.

Good safety and tolerability profiles

The most prevalent treatment-emergent adverse events (AEs) were fatigue, rash, cough, nausea, and decreased appetite, affecting 7-10 patients each. All were grade 1/2 except for one grade 3/4 case of fatigue and three cases of grade 3/4 anemia among the 23 patients. Most treatment-related AE’s were grade 1/2, largely fatigue (n = 8) and rash (n = 9). There was one case each of grade 3 adrenal insufficiency and hypokalemia. No patient discontinued the trial because of a treatment-related toxicity. Dr. Tolcher noted that adrenal insufficiency has been reported previously with the use of PD-1 inhibitors. “There does not appear to be any evidence of synergistic or additive toxicity in this patient population,” he said.

Neither drug affected the pharmacokinetics of the other drug or the development of antibodies to the other drug. The maximum tolerated dose of PF-2566 was at least 5 mg/kg every 3 weeks when combined with pembrolizumab 2 mg/kg. No dose-limiting toxicity was observed across the PF-2566 dosing range. And there were no treatment-emergent AEs of clinical relevance.

Pharmacodynamics and efficacy

By day 1 of cycle 5, “there [was] a trend toward increasing numbers of activated CD8 [cytotoxic] T cells in patients who ultimately responded or had a complete response, compared to those that had stable disease or progressive disease. The same actually applies to the effector memory T cells,” Dr. Tolcher said but was careful to point out that the sample sizes were small and it was only a trend. Similarly, circulating levels of gamma-interferon, often used as a biomarker of activated T cells, were higher at 6 and 24 hours post dose in cycle 5 for those patients who ultimately had partial or complete responses, compared with those with progressive or stable disease.

Among the 23 patients, there were two confirmed complete responses and four partial responses as well as one unconfirmed partial response. If responses occurred, they often were durable past 1 year and even out close to 2 years.

 

Dr. David Spigel

The strengths of this study were that it enrolled heavily pretreated patients and there were no drug-drug interactions, no dose-limiting toxicities, and no treatment-related AE’s leading to discontinuation, “so in general a very well-tolerated immunotherapy combination,” said discussant Dr. David Spigel of the Sarah Cannon Research Institute in Nashville, Tenn. There were also some durable responses, and he said it was interesting to see that there were some blood biomarkers that correlated with responses.

“It was hard for me to find any weaknesses to this,” Dr. Spigel said, beside the fact that it was a small study. “So what does this change?” He said the combination of pembrolizumab and PF-2566 looks promising in light of some sustained responses in refractory tumors and its safety profile. For the future, expansion trial cohorts are still needed to confirm activity and safety, especially hepatic safety based on trial results with similar drugs, and PF-2566 is already being tested with rituximab in lymphoma and with an anti-CCR4 compound (mogamulizumab).

 

 

The study was sponsored by Pfizer and Merck. Dr. Tolcher has ties to several companies, including Pfizer and Merck. Dr. Spigel has ties to several companies, including Pfizer.

Meeting/Event
Publications
Topics
Meeting/Event
Meeting/Event

CHICAGO – Combining an immunostimulatory agent with the PD-1 checkpoint inhibitor pembrolizumab appeared quite safe and very tolerable, in a small phase Ib study.

There were some signs of efficacy against a variety of solid tumors, as well as biomarker trends showing immune activity.

In the phase Ib trial, researchers combined escalating doses (0.45-5.0 mg/kg) of PF-2566, an investigative immunostimulatory agent, with the anti–PD-1 checkpoint inhibitor pembrolizumab at 2 mg/kg, with both drugs given intravenously once every 3 weeks for a maximum of 32 cycles. A primary objective of the trial was to determine a maximum tolerated dose. Secondary objectives were to assess safety and tolerability and to determine any antitumor responses.

 

Dr. Anthony Tolcher

PF-2566 (Utomilumab/PF-05082566) is a monoclonal agonist targeting 4-1BB, a “costimulatory molecule that’s induced upon T-cell receptor activation and ultimately enhances cytotoxic T-cell response and effector status,” said Dr. Anthony Tolcher of the START Center for Cancer Care, San Antonio, at the annual meeting of the American Society of Clinical Oncology.

Eligible patients were 18 years or older, had a performance status of 0-1, and had advanced or metastatic solid tumors that had progressed on standard therapy or for which no standard therapy was available. They could not have had any form of immunosuppressive therapy in the 2 weeks prior to registration, a monoclonal antibody in the 2 months before the first dose, or any symptomatic or progressing central nervous system primary malignancies. Prior pembrolizumab was permitted.

Twenty-three patients (14 males) were heavily pretreated with a median of three prior therapies (range 0-9) for a variety of cancers, including six non–small-cell lung, five renal cell, three head and neck, and two each pancreatic and thyroid cancers.

Good safety and tolerability profiles

The most prevalent treatment-emergent adverse events (AEs) were fatigue, rash, cough, nausea, and decreased appetite, affecting 7-10 patients each. All were grade 1/2 except for one grade 3/4 case of fatigue and three cases of grade 3/4 anemia among the 23 patients. Most treatment-related AE’s were grade 1/2, largely fatigue (n = 8) and rash (n = 9). There was one case each of grade 3 adrenal insufficiency and hypokalemia. No patient discontinued the trial because of a treatment-related toxicity. Dr. Tolcher noted that adrenal insufficiency has been reported previously with the use of PD-1 inhibitors. “There does not appear to be any evidence of synergistic or additive toxicity in this patient population,” he said.

Neither drug affected the pharmacokinetics of the other drug or the development of antibodies to the other drug. The maximum tolerated dose of PF-2566 was at least 5 mg/kg every 3 weeks when combined with pembrolizumab 2 mg/kg. No dose-limiting toxicity was observed across the PF-2566 dosing range. And there were no treatment-emergent AEs of clinical relevance.

Pharmacodynamics and efficacy

By day 1 of cycle 5, “there [was] a trend toward increasing numbers of activated CD8 [cytotoxic] T cells in patients who ultimately responded or had a complete response, compared to those that had stable disease or progressive disease. The same actually applies to the effector memory T cells,” Dr. Tolcher said but was careful to point out that the sample sizes were small and it was only a trend. Similarly, circulating levels of gamma-interferon, often used as a biomarker of activated T cells, were higher at 6 and 24 hours post dose in cycle 5 for those patients who ultimately had partial or complete responses, compared with those with progressive or stable disease.

Among the 23 patients, there were two confirmed complete responses and four partial responses as well as one unconfirmed partial response. If responses occurred, they often were durable past 1 year and even out close to 2 years.

 

Dr. David Spigel

The strengths of this study were that it enrolled heavily pretreated patients and there were no drug-drug interactions, no dose-limiting toxicities, and no treatment-related AE’s leading to discontinuation, “so in general a very well-tolerated immunotherapy combination,” said discussant Dr. David Spigel of the Sarah Cannon Research Institute in Nashville, Tenn. There were also some durable responses, and he said it was interesting to see that there were some blood biomarkers that correlated with responses.

“It was hard for me to find any weaknesses to this,” Dr. Spigel said, beside the fact that it was a small study. “So what does this change?” He said the combination of pembrolizumab and PF-2566 looks promising in light of some sustained responses in refractory tumors and its safety profile. For the future, expansion trial cohorts are still needed to confirm activity and safety, especially hepatic safety based on trial results with similar drugs, and PF-2566 is already being tested with rituximab in lymphoma and with an anti-CCR4 compound (mogamulizumab).

 

 

The study was sponsored by Pfizer and Merck. Dr. Tolcher has ties to several companies, including Pfizer and Merck. Dr. Spigel has ties to several companies, including Pfizer.

CHICAGO – Combining an immunostimulatory agent with the PD-1 checkpoint inhibitor pembrolizumab appeared quite safe and very tolerable, in a small phase Ib study.

There were some signs of efficacy against a variety of solid tumors, as well as biomarker trends showing immune activity.

In the phase Ib trial, researchers combined escalating doses (0.45-5.0 mg/kg) of PF-2566, an investigative immunostimulatory agent, with the anti–PD-1 checkpoint inhibitor pembrolizumab at 2 mg/kg, with both drugs given intravenously once every 3 weeks for a maximum of 32 cycles. A primary objective of the trial was to determine a maximum tolerated dose. Secondary objectives were to assess safety and tolerability and to determine any antitumor responses.

 

Dr. Anthony Tolcher

PF-2566 (Utomilumab/PF-05082566) is a monoclonal agonist targeting 4-1BB, a “costimulatory molecule that’s induced upon T-cell receptor activation and ultimately enhances cytotoxic T-cell response and effector status,” said Dr. Anthony Tolcher of the START Center for Cancer Care, San Antonio, at the annual meeting of the American Society of Clinical Oncology.

Eligible patients were 18 years or older, had a performance status of 0-1, and had advanced or metastatic solid tumors that had progressed on standard therapy or for which no standard therapy was available. They could not have had any form of immunosuppressive therapy in the 2 weeks prior to registration, a monoclonal antibody in the 2 months before the first dose, or any symptomatic or progressing central nervous system primary malignancies. Prior pembrolizumab was permitted.

Twenty-three patients (14 males) were heavily pretreated with a median of three prior therapies (range 0-9) for a variety of cancers, including six non–small-cell lung, five renal cell, three head and neck, and two each pancreatic and thyroid cancers.

Good safety and tolerability profiles

The most prevalent treatment-emergent adverse events (AEs) were fatigue, rash, cough, nausea, and decreased appetite, affecting 7-10 patients each. All were grade 1/2 except for one grade 3/4 case of fatigue and three cases of grade 3/4 anemia among the 23 patients. Most treatment-related AE’s were grade 1/2, largely fatigue (n = 8) and rash (n = 9). There was one case each of grade 3 adrenal insufficiency and hypokalemia. No patient discontinued the trial because of a treatment-related toxicity. Dr. Tolcher noted that adrenal insufficiency has been reported previously with the use of PD-1 inhibitors. “There does not appear to be any evidence of synergistic or additive toxicity in this patient population,” he said.

Neither drug affected the pharmacokinetics of the other drug or the development of antibodies to the other drug. The maximum tolerated dose of PF-2566 was at least 5 mg/kg every 3 weeks when combined with pembrolizumab 2 mg/kg. No dose-limiting toxicity was observed across the PF-2566 dosing range. And there were no treatment-emergent AEs of clinical relevance.

Pharmacodynamics and efficacy

By day 1 of cycle 5, “there [was] a trend toward increasing numbers of activated CD8 [cytotoxic] T cells in patients who ultimately responded or had a complete response, compared to those that had stable disease or progressive disease. The same actually applies to the effector memory T cells,” Dr. Tolcher said but was careful to point out that the sample sizes were small and it was only a trend. Similarly, circulating levels of gamma-interferon, often used as a biomarker of activated T cells, were higher at 6 and 24 hours post dose in cycle 5 for those patients who ultimately had partial or complete responses, compared with those with progressive or stable disease.

Among the 23 patients, there were two confirmed complete responses and four partial responses as well as one unconfirmed partial response. If responses occurred, they often were durable past 1 year and even out close to 2 years.

 

Dr. David Spigel

The strengths of this study were that it enrolled heavily pretreated patients and there were no drug-drug interactions, no dose-limiting toxicities, and no treatment-related AE’s leading to discontinuation, “so in general a very well-tolerated immunotherapy combination,” said discussant Dr. David Spigel of the Sarah Cannon Research Institute in Nashville, Tenn. There were also some durable responses, and he said it was interesting to see that there were some blood biomarkers that correlated with responses.

“It was hard for me to find any weaknesses to this,” Dr. Spigel said, beside the fact that it was a small study. “So what does this change?” He said the combination of pembrolizumab and PF-2566 looks promising in light of some sustained responses in refractory tumors and its safety profile. For the future, expansion trial cohorts are still needed to confirm activity and safety, especially hepatic safety based on trial results with similar drugs, and PF-2566 is already being tested with rituximab in lymphoma and with an anti-CCR4 compound (mogamulizumab).

 

 

The study was sponsored by Pfizer and Merck. Dr. Tolcher has ties to several companies, including Pfizer and Merck. Dr. Spigel has ties to several companies, including Pfizer.

Publications
Publications
Topics
Article Type
Display Headline
Pembrolizumab paired with immunostimulator is safe and tolerable
Display Headline
Pembrolizumab paired with immunostimulator is safe and tolerable
Article Source

AT THE 2016 ASCO ANNUAL MEETING

Disallow All Ads
Vitals

Key clinical point: Combining an immunostimulator with pembrolizumab had good tolerability and safety.

Major finding: Two complete and four partial responses occurred among 23 patients.

Data source: Phase Ib trial of 23 patients with a variety of solid tumors.

Disclosures: The study was sponsored by Pfizer and Merck. Dr. Tolcher has ties to several companies, including Pfizer and Merck. Dr. Spigel has ties to several companies, including Pfizer.

Neoadjuvant chemo for advanced ovarian cancer opens ‘window of opportunity’ for immunotherapies

Article Type
Changed
Fri, 01/04/2019 - 13:17
Display Headline
Neoadjuvant chemo for advanced ovarian cancer opens ‘window of opportunity’ for immunotherapies

Presurgical neoadjuvant chemotherapy (NACT) altered the immune microenvironment in patients with stage IIIC/IV tubo-ovarian high-grade serous carcinoma (HGSC), perhaps enhancing the potential response to immunotherapy, investigators report.

“The results suggest that the effects of immunotherapy might be enhanced if given after chemotherapy, potentially improving disease control in patients with advanced HGSC and other cancer types,” wrote Dr. Steffen Bohm of Barts Cancer Institute in London and his associates (CCR. 2016. doi: 10.1158/1078-0432.CCR-15-2657).

Dr. Bohm and associates collected pre- and postchemotherapy biopsies and blood samples from 60 patients with stage IIIC or IV HGSC; 54 patients underwent platinum-based neoadjuvant chemotherapy and 6 received chemotherapy after debulking surgery. The investigators used immunohistochemistry and RNA sequencing to evaluate the changes before and after chemotherapy. The patients were grouped by their response to chemotherapy.

Results indicated that NACT induced T cell activation, with results most pronounced among those who had a good response to chemotherapy. Among a subset of 25 patients, omental metastases biopsies indicated CD8+ T cells and memory cells were present in the tumors after NACT. Proinflammatory cytokines decreased in all patients following NACT.

Immunohistochemical staining for PD-1 ligands showed that PD-L1 levels were significantly increased in post-NACT samples regardless of a patient’s response to chemotherapy.

“Our results suggest that NACT opens a window of opportunity for immunotherapies such as immune checkpoint blockade for patients with different levels of response to chemotherapy,” Dr. Bohm and associates said.

The study was funded by the Swiss Cancer League, the European Research Council, Cancer Research UK, and Barts and The London Charity. The authors had no relevant disclosures to report.

[email protected]

On Twitter @jessnicolecraig

Publications
Topics
Sections

Presurgical neoadjuvant chemotherapy (NACT) altered the immune microenvironment in patients with stage IIIC/IV tubo-ovarian high-grade serous carcinoma (HGSC), perhaps enhancing the potential response to immunotherapy, investigators report.

“The results suggest that the effects of immunotherapy might be enhanced if given after chemotherapy, potentially improving disease control in patients with advanced HGSC and other cancer types,” wrote Dr. Steffen Bohm of Barts Cancer Institute in London and his associates (CCR. 2016. doi: 10.1158/1078-0432.CCR-15-2657).

Dr. Bohm and associates collected pre- and postchemotherapy biopsies and blood samples from 60 patients with stage IIIC or IV HGSC; 54 patients underwent platinum-based neoadjuvant chemotherapy and 6 received chemotherapy after debulking surgery. The investigators used immunohistochemistry and RNA sequencing to evaluate the changes before and after chemotherapy. The patients were grouped by their response to chemotherapy.

Results indicated that NACT induced T cell activation, with results most pronounced among those who had a good response to chemotherapy. Among a subset of 25 patients, omental metastases biopsies indicated CD8+ T cells and memory cells were present in the tumors after NACT. Proinflammatory cytokines decreased in all patients following NACT.

Immunohistochemical staining for PD-1 ligands showed that PD-L1 levels were significantly increased in post-NACT samples regardless of a patient’s response to chemotherapy.

“Our results suggest that NACT opens a window of opportunity for immunotherapies such as immune checkpoint blockade for patients with different levels of response to chemotherapy,” Dr. Bohm and associates said.

The study was funded by the Swiss Cancer League, the European Research Council, Cancer Research UK, and Barts and The London Charity. The authors had no relevant disclosures to report.

[email protected]

On Twitter @jessnicolecraig

Presurgical neoadjuvant chemotherapy (NACT) altered the immune microenvironment in patients with stage IIIC/IV tubo-ovarian high-grade serous carcinoma (HGSC), perhaps enhancing the potential response to immunotherapy, investigators report.

“The results suggest that the effects of immunotherapy might be enhanced if given after chemotherapy, potentially improving disease control in patients with advanced HGSC and other cancer types,” wrote Dr. Steffen Bohm of Barts Cancer Institute in London and his associates (CCR. 2016. doi: 10.1158/1078-0432.CCR-15-2657).

Dr. Bohm and associates collected pre- and postchemotherapy biopsies and blood samples from 60 patients with stage IIIC or IV HGSC; 54 patients underwent platinum-based neoadjuvant chemotherapy and 6 received chemotherapy after debulking surgery. The investigators used immunohistochemistry and RNA sequencing to evaluate the changes before and after chemotherapy. The patients were grouped by their response to chemotherapy.

Results indicated that NACT induced T cell activation, with results most pronounced among those who had a good response to chemotherapy. Among a subset of 25 patients, omental metastases biopsies indicated CD8+ T cells and memory cells were present in the tumors after NACT. Proinflammatory cytokines decreased in all patients following NACT.

Immunohistochemical staining for PD-1 ligands showed that PD-L1 levels were significantly increased in post-NACT samples regardless of a patient’s response to chemotherapy.

“Our results suggest that NACT opens a window of opportunity for immunotherapies such as immune checkpoint blockade for patients with different levels of response to chemotherapy,” Dr. Bohm and associates said.

The study was funded by the Swiss Cancer League, the European Research Council, Cancer Research UK, and Barts and The London Charity. The authors had no relevant disclosures to report.

[email protected]

On Twitter @jessnicolecraig

Publications
Publications
Topics
Article Type
Display Headline
Neoadjuvant chemo for advanced ovarian cancer opens ‘window of opportunity’ for immunotherapies
Display Headline
Neoadjuvant chemo for advanced ovarian cancer opens ‘window of opportunity’ for immunotherapies
Sections
Article Source

FROM CLINICAL CANCER RESEARCH

Disallow All Ads
Vitals

Key clinical point: Neoadjuvant chemotherapy alters the immune microenvironment in patients with advanced ovarian cancer.

Major finding: Neoadjuvant chemotherapy induced T cell activity in patients with stage IIIC/IV tubo-ovarian high-grade serous carcinoma. Proinflammatory cytokines decreased in all patients following neoadjuvant chemotherapy.

Data source: A longitudinal cohort study of 60 patients with stage IIIC/IV tubo-ovarian high-grade serous carcinoma.

Disclosures: The research was funded by the Swiss Cancer League, the European Research Council, Cancer Research UK, and Barts and The London Charity. The authors had no relevant disclosures to report.

Will VA & DoD Be Part of the Cancer Moon Shot?

Article Type
Changed
Thu, 12/15/2022 - 14:59
Display Headline
Will VA & DoD Be Part of the Cancer Moon Shot?
White House promises that the federal government will do everything it possibly can   to improve cancer care through funding, targeted incentives, and increased private-sector coordination.

Updated January 20, 2016

The Million Veteran Program may play an important role in the recently announced cancer "moon shot." President Barack Obama and Vice President Joe Biden are promising to marshal federal and private resources to battle against cancer. Beginning with a bold announcement at the President’s State of the Union address, delivered January 12, and followed up by a slightly more detailed plan put forth by Biden a day later, the White House is focused on battling cancer by breaking down silos and increasing both public and private resources.

“The goal of this initiative — this ‘Moonshot’ — is to seize this moment,” Biden explained in a blog post. “To accelerate our efforts to progress towards a cure, and to unleash new discoveries and breakthroughs for other deadly diseases.”

Noting advances in immunotherapy, genomics and combined therapies, Biden argued that great strides have been made in cancer research, but the results are not necessarily reaching patients. “The science, data, and research results are trapped in silos, preventing faster progress and greater reach to patients,” Biden insisted. “It’s not just about developing game-changing treatments — it’s about delivering them to those who need them.”

Biden pledged that the federal government “will do everything it possibly can— through funding, targeted incentives, and increased private-sector coordination — to support research and enable progress.” Unclear, however, is whether the VA or the Murtha Cancer Center at Walter Reed National Military Medical Center will be involved in the effort. Later this month Biden will meet with cabinet secretaries and heads of relevant agencies to discuss ways to improve federal investment and support of cancer research and treatment.

VA Secretary Bob McDonald recently toured a Million Veteran Program (MVP) repository in Boston and touted the potential role in could play in the cancer moon shot, according to Military.com. Veterans who participate in the program donate blood for DNA extraction, which is linked to their health records. Created in 2012, MVP was expected to take 5 to 7 years to reach 1 million participants and already has registered more than 400,000 participants.

According to Politico, Biden has already made significant progress. The White House has a detailed plan leveraging the work of the National Institutes of Health (NIH) and private organizations that can compress 10 years of work into 5. The NIH is one of the few agencies that has received more funding from Congress in the latest budgets.

Medical and cancer organizations met the announcement with widespread approval. The American Association for Cancer Research (AACR) applauded the commitment to curing cancer. “We have indeed reached an inflection point, where the number of discoveries that are being made at such an accelerated pace are saving lives and bringing enormous hope for cancer patients, even those with advanced disease,” Dr. José Baselga, AACR president, said in a statement. “Now is the time for a major new initiative in cancer science that supports and builds upon our basic science foundation while translating these exciting scientific discoveries into improved treatments for cancer patients, such as in the areas of genomics, precision medicine, and immuno-oncology.”

“Vice President Biden’s call to leading cancer centers to break down silos and reach unprecedented levels of cooperation to enhance the effectiveness of cancer treatment, and for the oncology community to improve communication so that the care provided to patients at the world’s best cancer centers is available to everyone who needs it, echoes the work and mission of NCCN and our Member Institutions,” said Robert W. Carlson, MD, chief executive officer of the National Comprehensive Cancer Network (NCCN). “NCCN stands with President Obama, Vice President Biden, and their Administration on this crucial initiative, and we look forward to working to advance the goals of the initiative. It is time that people stop dying of cancer.”

The American Society of Clinical Oncology (ASCO) concurred. “With nearly 1.7 million people in the United States diagnosed with cancer each year, and the incidence of cancer expected to rise to 2.3 million cases per year by 2030, it is imperative that we do all we can to bring more effective treatments from the laboratory bench to the patient’s bedside as quickly as possible,”  Richard L. Schilsky, MD, ASCO chief medical officer, said in a statement. “We must recommit to vastly speeding the discovery of new cancer treatments and enabling the possibility of precision medicine for every individual with cancer.”

Publications
Topics
Legacy Keywords
cancer, President Barack Obama, Vice President Joe Biden, National Institutes of Health, The American Association for Cancer Research,
Sections
Related Articles
White House promises that the federal government will do everything it possibly can   to improve cancer care through funding, targeted incentives, and increased private-sector coordination.
White House promises that the federal government will do everything it possibly can   to improve cancer care through funding, targeted incentives, and increased private-sector coordination.

Updated January 20, 2016

The Million Veteran Program may play an important role in the recently announced cancer "moon shot." President Barack Obama and Vice President Joe Biden are promising to marshal federal and private resources to battle against cancer. Beginning with a bold announcement at the President’s State of the Union address, delivered January 12, and followed up by a slightly more detailed plan put forth by Biden a day later, the White House is focused on battling cancer by breaking down silos and increasing both public and private resources.

“The goal of this initiative — this ‘Moonshot’ — is to seize this moment,” Biden explained in a blog post. “To accelerate our efforts to progress towards a cure, and to unleash new discoveries and breakthroughs for other deadly diseases.”

Noting advances in immunotherapy, genomics and combined therapies, Biden argued that great strides have been made in cancer research, but the results are not necessarily reaching patients. “The science, data, and research results are trapped in silos, preventing faster progress and greater reach to patients,” Biden insisted. “It’s not just about developing game-changing treatments — it’s about delivering them to those who need them.”

Biden pledged that the federal government “will do everything it possibly can— through funding, targeted incentives, and increased private-sector coordination — to support research and enable progress.” Unclear, however, is whether the VA or the Murtha Cancer Center at Walter Reed National Military Medical Center will be involved in the effort. Later this month Biden will meet with cabinet secretaries and heads of relevant agencies to discuss ways to improve federal investment and support of cancer research and treatment.

VA Secretary Bob McDonald recently toured a Million Veteran Program (MVP) repository in Boston and touted the potential role in could play in the cancer moon shot, according to Military.com. Veterans who participate in the program donate blood for DNA extraction, which is linked to their health records. Created in 2012, MVP was expected to take 5 to 7 years to reach 1 million participants and already has registered more than 400,000 participants.

According to Politico, Biden has already made significant progress. The White House has a detailed plan leveraging the work of the National Institutes of Health (NIH) and private organizations that can compress 10 years of work into 5. The NIH is one of the few agencies that has received more funding from Congress in the latest budgets.

Medical and cancer organizations met the announcement with widespread approval. The American Association for Cancer Research (AACR) applauded the commitment to curing cancer. “We have indeed reached an inflection point, where the number of discoveries that are being made at such an accelerated pace are saving lives and bringing enormous hope for cancer patients, even those with advanced disease,” Dr. José Baselga, AACR president, said in a statement. “Now is the time for a major new initiative in cancer science that supports and builds upon our basic science foundation while translating these exciting scientific discoveries into improved treatments for cancer patients, such as in the areas of genomics, precision medicine, and immuno-oncology.”

“Vice President Biden’s call to leading cancer centers to break down silos and reach unprecedented levels of cooperation to enhance the effectiveness of cancer treatment, and for the oncology community to improve communication so that the care provided to patients at the world’s best cancer centers is available to everyone who needs it, echoes the work and mission of NCCN and our Member Institutions,” said Robert W. Carlson, MD, chief executive officer of the National Comprehensive Cancer Network (NCCN). “NCCN stands with President Obama, Vice President Biden, and their Administration on this crucial initiative, and we look forward to working to advance the goals of the initiative. It is time that people stop dying of cancer.”

The American Society of Clinical Oncology (ASCO) concurred. “With nearly 1.7 million people in the United States diagnosed with cancer each year, and the incidence of cancer expected to rise to 2.3 million cases per year by 2030, it is imperative that we do all we can to bring more effective treatments from the laboratory bench to the patient’s bedside as quickly as possible,”  Richard L. Schilsky, MD, ASCO chief medical officer, said in a statement. “We must recommit to vastly speeding the discovery of new cancer treatments and enabling the possibility of precision medicine for every individual with cancer.”

Updated January 20, 2016

The Million Veteran Program may play an important role in the recently announced cancer "moon shot." President Barack Obama and Vice President Joe Biden are promising to marshal federal and private resources to battle against cancer. Beginning with a bold announcement at the President’s State of the Union address, delivered January 12, and followed up by a slightly more detailed plan put forth by Biden a day later, the White House is focused on battling cancer by breaking down silos and increasing both public and private resources.

“The goal of this initiative — this ‘Moonshot’ — is to seize this moment,” Biden explained in a blog post. “To accelerate our efforts to progress towards a cure, and to unleash new discoveries and breakthroughs for other deadly diseases.”

Noting advances in immunotherapy, genomics and combined therapies, Biden argued that great strides have been made in cancer research, but the results are not necessarily reaching patients. “The science, data, and research results are trapped in silos, preventing faster progress and greater reach to patients,” Biden insisted. “It’s not just about developing game-changing treatments — it’s about delivering them to those who need them.”

Biden pledged that the federal government “will do everything it possibly can— through funding, targeted incentives, and increased private-sector coordination — to support research and enable progress.” Unclear, however, is whether the VA or the Murtha Cancer Center at Walter Reed National Military Medical Center will be involved in the effort. Later this month Biden will meet with cabinet secretaries and heads of relevant agencies to discuss ways to improve federal investment and support of cancer research and treatment.

VA Secretary Bob McDonald recently toured a Million Veteran Program (MVP) repository in Boston and touted the potential role in could play in the cancer moon shot, according to Military.com. Veterans who participate in the program donate blood for DNA extraction, which is linked to their health records. Created in 2012, MVP was expected to take 5 to 7 years to reach 1 million participants and already has registered more than 400,000 participants.

According to Politico, Biden has already made significant progress. The White House has a detailed plan leveraging the work of the National Institutes of Health (NIH) and private organizations that can compress 10 years of work into 5. The NIH is one of the few agencies that has received more funding from Congress in the latest budgets.

Medical and cancer organizations met the announcement with widespread approval. The American Association for Cancer Research (AACR) applauded the commitment to curing cancer. “We have indeed reached an inflection point, where the number of discoveries that are being made at such an accelerated pace are saving lives and bringing enormous hope for cancer patients, even those with advanced disease,” Dr. José Baselga, AACR president, said in a statement. “Now is the time for a major new initiative in cancer science that supports and builds upon our basic science foundation while translating these exciting scientific discoveries into improved treatments for cancer patients, such as in the areas of genomics, precision medicine, and immuno-oncology.”

“Vice President Biden’s call to leading cancer centers to break down silos and reach unprecedented levels of cooperation to enhance the effectiveness of cancer treatment, and for the oncology community to improve communication so that the care provided to patients at the world’s best cancer centers is available to everyone who needs it, echoes the work and mission of NCCN and our Member Institutions,” said Robert W. Carlson, MD, chief executive officer of the National Comprehensive Cancer Network (NCCN). “NCCN stands with President Obama, Vice President Biden, and their Administration on this crucial initiative, and we look forward to working to advance the goals of the initiative. It is time that people stop dying of cancer.”

The American Society of Clinical Oncology (ASCO) concurred. “With nearly 1.7 million people in the United States diagnosed with cancer each year, and the incidence of cancer expected to rise to 2.3 million cases per year by 2030, it is imperative that we do all we can to bring more effective treatments from the laboratory bench to the patient’s bedside as quickly as possible,”  Richard L. Schilsky, MD, ASCO chief medical officer, said in a statement. “We must recommit to vastly speeding the discovery of new cancer treatments and enabling the possibility of precision medicine for every individual with cancer.”

Publications
Publications
Topics
Article Type
Display Headline
Will VA & DoD Be Part of the Cancer Moon Shot?
Display Headline
Will VA & DoD Be Part of the Cancer Moon Shot?
Legacy Keywords
cancer, President Barack Obama, Vice President Joe Biden, National Institutes of Health, The American Association for Cancer Research,
Legacy Keywords
cancer, President Barack Obama, Vice President Joe Biden, National Institutes of Health, The American Association for Cancer Research,
Sections
Disallow All Ads
Alternative CME

Harnessing Vaccines to Treat Cancers

Article Type
Changed
Thu, 12/15/2022 - 15:01
Display Headline
Harnessing Vaccines to Treat Cancers
Therapeutic vaccines promise new and potentially more effective treatment options for solid tumors and hematologic malignancies.

Vaccines are used to prevent bacterial infections, such as pneumonia, and viral diseases, such as influenza.1 Recently, they are being used to prevent cancers. For example, the human papilloma virus vaccine prevents infection with the virus that is associated with cervical cancer.2 Now there is another purpose for vaccines: They can be used therapeutically. Vaccines can be given even after a person manifests a tumor; they can cause the tumor to shrink or disappear. Therapeutic vaccines hold the potential to effect profound changes in the treatment of several cancers.

Biologics

Biologics, a recently developed class of weapons, can be but aren’t always relatively specific to cancer cells. Biologic agents include inhibitors and monoclonal antibodies. Inhibitors target specific functions, such as angiogenesis. However, inhibitors are not always selective for cancer cells. Angiogenesis inhibitors block formation of blood vessels. Like cytotoxic chemotherapy agents, they block the formation of blood vessels in normal cells as well as cancer cells. Cancer cells, however, generally grow more rapidly than do normal host cells, so they need more blood vessels faster to nourish their cells.3

Some inhibitors are more specific. An inhibitor molecule can be directed against a specific transcript that does not occur in normal cells. In normal cells, the breakpoint cluster region (BCR) gene on chromosome 22 directs synthesis of its protein product, whereas the abelson murine leukemia viral oncogene homolog 1 (ABL1) gene on chromosome 9 specifies another protein. However, in chronic myelogenous leukemia, the ends of those 2 chromosomes translocate, and the BCR/ABL1 transcript is a hybrid of RNA derived from the original chromosome and that of the newly attached, translocated chromosome 9. This hybrid RNA directs synthesis of a fusion protein, a tyrosine kinase; the fusion protein is not produced in normal host cells. Thus imatinib, an inhibitor directed at the fusion protein, inhibits the cancer cells specifically.4

Monoclonal Antibodies

Monoclonal antibodies exploit the host’s immune system to destroy cancer cells. Like the inhibitors, monoclonal antibodies can be directed against a specific functional protein, which is not necessarily specific to tumor cells, or against a protein or family of proteins unique to the cancer cells. For example, trastuzumab is a monoclonal antibody directed against the HER2/neu oncogene, which is amplified in some breast cancers.5 The host’s immune system recognizes the antibodyantigen complex and signals its macrophages to destroy the complex, and the cancer cell dies.

Vaccines also exploit the immune system. One way to immunize against a virus, is to introduce a live or killed virus or a part of a virus—usually a part of a viral protein—into the recipient. The immune system recognizes the foreign virus and makes antibodies. When the immune system is challenged by exposure to the pathogenic virus, the antiviral antibodies recognize and bind to it. The host’s macrophages then engulf the antigenantibody complex and destroy it.6 This type of vaccine prevents infection.

Viral Vaccines

Few vaccines prevent specific cancers. Immunization against the hepatitis B virus, for example, confers immunity to a virus whose infection is a major risk factor for hepatocellular carcinoma.7 Thus it is not truly an anticancer vaccine but rather an antiviral vaccine. Similarly, the HPV
vaccine does not prevent cervical cancer but confers immunity
to a virus that causes cervical cancer.2

Using vaccines to prevent cancer in general is now becoming feasible. Cancer vaccines in use or in development at this time are of 2 general types: those derived from a single tumor and are designed to elicit immunity to that particular tumor in that particular individual host; and those designed to provoke an immune response to that particular type of tumor in any host. The latter type can be mass produced, whereas vaccines of the first type are restricted to the host of origin.

Autologous Vaccines

At the time of biopsy or surgical excision, tumors are mechanically and enzymatically dissociated into single cells, then grown in tissue culture. When they are cultured in suspension with agitation, the cancer stem cells (CSCs) form spheroids, and other cell types do not. The CSCs are the progenitor cells of the specific tumor, much like bone marrow stem cells are the progenitors of the myeloid, erythroid, and megakaryocyte (platelet) cell lineages of human blood. After the CSCs are expanded, the spheroids are harvested, and their RNA is extracted. The messenger RNAs, which specify the proteins made by the CSCs, are converted to complementary DNAs, which are then amplified using the polymerase chain reaction.8

Meanwhile, the patient’s peripheral blood mononuclear cells (PBMCs) are harvested from the blood by leukapheresis. The PBMCs are then enriched for monocytes by immunologic depletion of B and T cells. The monocytes are cultured in the presence of interleukin-4 and granulocytemacrophage-colony-stimulating factor (GM-CSF), and they become immature dendritic cells after 5 days. These immature dendritic cells are transfected with the amplified DNA from the CSC tumor spheres and grown for 2 more days in a medium supplemented with interleukin-1b (IL-1b), interleukin-6 (IL-6), tumor necrosis factor-α, and prostaglandin E2, then tested for markers characteristic of dendritic cells and frozen.

Aliquots of these transfected dendritic cells are injected into the patient at intervals over several months. Dendritic cells, which are the most efficient cells at eliciting an immune response, present the tumor CSC antigens to the patient’s immune system, which develops antibodies to the antigens presented on the dendritic cells and proceeds to destroy the tumor CSCs remaining in the patient’s tumor. Without its CSCs, the tumor is unable to revitalize itself, the rest of the tumor cells eventually die, and the tumor shrinks. The patient is monitored for immune response, and the tumor is regularly imaged. This vaccine is an example of therapeutic use: It can cause tumor regression.8

Sipuleucil-T is another example of an autologous vaccine used for therapeutic purposes. The patient’s antigenpresenting cells (APCs) are harvested by leukopheresis. They are then cultured in the presence of the protein made by fusing prostatic antigen phosphatase (PAP) with the GM-CSF to form the fusion protein PAP-GM-CSF. These modified APCs are then reinjected into the same host. Sipuleucil-T is now FDA approved for the treatment of metastatic castrate-resistant prostate cancer (mCRPC) and has been shown to extend life by approximately 4 months.9

Universal Vaccine for a Specific Tumor

Cancer vaccines can also be prepared in a more conventional manner, analogous to the more commonly used vaccines, and given to all patients with that specific tumor. PROSTVAC is another vaccine for mCRPC as well as for earlier-stage prostate cancer. The DNA specifying prostate-specific antigen (PSA), the secreted protein expressed by the tumor, is mutated very slightly and inserted into a vaccinia virus (Figure). Also inserted into the vaccinia virus are DNAs encoding 3 proteins that
stimulate the patient’s immune system: B7.1, intercellular adhesion molecule-1 (ICAM-1), and leukocyte functionassociated antigen-3 (LFA-3).

This recombinant viral vector is propagated in tissue culture, purified, and administered to the patient subcutaneously. The virus replicates for a short period in the host, expressing the 3 immunostimulatory molecules and the mutated PSA molecule protein product, which differs by 1 amino acid from that expressed by the patient’s tumor. The vaccinia virus and the mutant protein are recognized as foreign by the patient’s immune system, whereas the 3 immunostimulatory molecules are not, as they are the same as the host’s immunostimulatory molecules.

Subsequent boosts, administered over about 5 months, are with a fowlpox virus vector (which does not replicate in the patient) containing the same 4 genes that had been inserted into the vaccinia virus. This strategy enhances the immune response to the mutant tumor product and ensures that the host is not generating a response to the vaccinia virus. The mutant tumor product is sufficiently similar to the patient’s native tumor product that the immune system mounts a response to both proteins. The T cells generated in response to the mutant protein bind to both PSA and mutated PSA molecule proteins, and cells harboring these proteins are destroyed.10

Whereas the T-cell response is important for the activity of the PROSTVAC vaccine, and there is no evidence for formation of anti-PSA antibodies, other vaccines exploit both B- and T-cell responses.10,11 After a virus commandeers the host cell’s replication, transcription, and translation machinery, the macrophage breaks down the viral protein product (the mutated or immunogenic molecule) into small fragments. These fragments bind to major histocompatibility complex (MHC) class II molecules, which are produced by the macrophages. These complexes of antigen (virus) fragments with MHC class II molecules are transported to the surface of the macrophage, where the antigen is presented to lymphocytes. A receptor on a B lymphocyte adheres to the viral antigen and thus becomes an activated B cell, which divides and produces many copies. The B lymphocytes mature into plasma cells and release antibodies that adhere to the virus. A helper T-cell receptor can adhere to the antigen-MHC class II complex displayed on the B cell and activate the lymphocyte, causing the release of more and different cytokines. The cytokines stimulate the activated B cell to divide, producing functionally mature antibodies that recognize and attach to the virus. Macrophages can then engulf and destroy the virus, or the antibody-linked viruses can be excreted in the urine or stool.12

When viruses (eg, the recombinant vaccinia virus of the PROSTVAC vaccine) infect cells and replicate, fragments of viral proteins become attached to MHC class I molecules (see Box). These complexes attach to the cell surface and are presented to cytotoxic T cells. The activated cytotoxic T cells divide and destroy cells harboring the virus or viral antigens.12

Some of the B lymphocytes become memory B cells. Similarly, some of the T lymphocytes become memory T cells. These memory B cells respond and replicate rapidly on re-exposure to the virus or viral proteins to produce antigen-specific antibodies.12

When a macrophage engulfs and breaks down a tumor cell harboring a PSA molecule, other tumor-specific protein fragments are released. These tumor-specific fragments then serve as antigens and elicit an immune response, generating even more antitumor antibodies and T-cell interactions. Thus the tumor regresses. Importantly, this type of vaccine can be mass produced.

Vaccines With Chemotherapy

Another approach to the design of a tumor-specific vaccine is based on tumor-associated peptides (TUMAPs). These TUMAPs are highly overexpressed in tumors relative to a number of normal, healthy tissues. Nine highly immunogenic TUMAPs from 80 specimens of the same tumor type, identified by mass spectrometry, gene expression profiling, literature-based functional assessment, bioinformatics, and human T-cell assays, are used to prepare IMA901, a multipeptide vaccine against the specific tumor, renal cell carcinoma. The pool of selected peptides, composed of 9 to 16 amino acids, is then administered to patients following a dose of the immunomodulator GM-CSF for a total of as many as 17 injections. Low-dose cyclophosphamide administered prior to the first vaccination downregulates regulator T cells, enhancing immune response and resulting in prolonged survival.13 This type of vaccine can also be mass produced.

Conclusions

The vaccines that are currently in use or in clinical trials are being used in patients with advanced disease. As such, they are therapeutic rather than prophylactic. It is inevitable that more vaccines will earn FDA approval. Eventually, they will be used in earlier-stage disease. It is conceivable that some can be used prophylactically when a malignancy first becomes detectable, especially as more sensitive detection methods are developed. Vaccine delivery systems are likely to change as researchers investigate and adopt matrix materials that increase efficacy.

Cancer vaccines hold the potential for greater selectivity in treatment of malignancies. These vaccines may enable us to use cytotoxic chemotherapy agents less often or perhaps more effectively when used in conjunction with vaccines. Vaccines that can be mass produced could conceivably decrease the financial burden of cancer treatment as well as the human cost of malignant diseases and their therapy and care requirements.

Solid-tumor and hematologic malignancy vaccines are coming into wider use as the science is better understood and more methods of generating immune responses are explored. A personalized cancer vaccine, sipuleucel-T, is FDA approved for clinical use against mCRPC.9 A poxvirus-based vaccine, PROSTVAC, also against mCRPC, is in phase 3 clinical trials.10,11 The multipeptide renal cell carcinoma vaccine IMA901 is also in phase 3 clinical trials.13 A personalized vaccine against glioblastoma multiforme CSCs has extended progression-free survival in its initial pretrial study.8,14 Additional glioblastoma, melanoma, and other vaccines are in clinical trials and under development.

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Click here to continue reading.

References

1. U.S. Department of Health and Human Services, National Institutes of Health, National Institute of Allergy and Infectious Diseases. Understanding vaccines: what they are, how they work. National Institute of Allergy and Infectious Diseases Website. http://www.niaid.nih.gov/topics/vaccines/documents/undvacc.pdf. Published January 2008. Accessed March 24, 2015. .

2. Hagensee ME, Yaegashi N, Galloway GA. Self-assembly of human papilloma virus type 1 capsids by expression of the L1 protein alone or by coexpression of the L1 and L2 capsid proteins. J Virol. 1993;67(1):315-322.

3. American Cancer Society. Anti-angiogenesis treatment. American Cancer Society Website. http://www.cancer.org/acs/groups/cid/documents/webcontent/002988-pdf.pdf. Revised March 10, 2009. Accessed March 24, 2015.

4. Gambacorti-Passerini CB, Gunby RH, Piazza R, Galietta A, Rostagno R, Scapozza L. Molecular mechanisms of resistance to imatinib in Philadelphia-chromosomepositive leukaemias. Lancet Oncol. 2003;4(2):75-85.

5. Hudis CA. Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39-51.

6. NPI reference guide on vaccines and vaccine safety: How vaccines work. PATH Website. http://www.path.org/vaccineresources/files/How_Vaccines_Work.pdf. Accessed March 24, 2015.

7. Beasley RP, Hwang LY, Lin CC, Chien CS. Hepatocellular carcinoma and hepatitis B virus: a prospective study of 22 707 men in Taiwan. Lancet. 1981;318(8256):1129-1133.

8. Vik-Mo EO, Nyakas M, Mikkelsen BV, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62(9):1499-1509.

9. Kantoff PW, Higano CS, Shore ND, et al; IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411-422.

10. Gulley JL, Madan RA, Tsang KY, et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res. 2014;2(2):133-141.

11. Campbell CT, Gulley JL, Oyelaran O, Hodge JW, Schlom J, Gildersleeve JC. Humoral response to a viral glycan correlates with survival on PROSTVAC VF. Proc Natl Acad Sci USA. 2014;111(17):E1749-E1758.

12. Public Health England. Immunity and how vaccines work: The green book, chapter 1. GOV.UK Website. https://www.gov.uk/government/publications/immunity-and-how-vaccines-work-the-green-book-chapter-1. Published March 19, 2013. Accessed March 24, 2015.

13. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18(8):1254-1261.

14. Ottenhausen M, Bodhinayake I, Banu M, Kesavabhotla K, Ray A, Boockvar JA. Industry progress report on neuro-oncology: biotech update 2013. J Neurooncol. 2013;115(2):311-316.

Author and Disclosure Information

Dr. Kuemmerle is a staff physician in the Section of Hematology and Oncology at the White River Junction VAMC in Vermont. She is also an assistant professor of medicine at the Geisel School of Medicine at Dartmouth in Hanover, New Hampshire

Issue
Federal Practitioner - 32(4)s
Publications
Topics
Page Number
27S-30S
Legacy Keywords
cancer vaccines, biologics, biologic agents, angiogenesis inhibitors, monoclonal antibodies, viral vaccines, autologous vaccines, poxvirus vector, PROSTVAC, vaccines with chemotherapy, tumor-specific vaccine, tumor-associated peptides, TUMAPs, sipuleucel-T personalized cancer vaccine
Sections
Author and Disclosure Information

Dr. Kuemmerle is a staff physician in the Section of Hematology and Oncology at the White River Junction VAMC in Vermont. She is also an assistant professor of medicine at the Geisel School of Medicine at Dartmouth in Hanover, New Hampshire

Author and Disclosure Information

Dr. Kuemmerle is a staff physician in the Section of Hematology and Oncology at the White River Junction VAMC in Vermont. She is also an assistant professor of medicine at the Geisel School of Medicine at Dartmouth in Hanover, New Hampshire

Therapeutic vaccines promise new and potentially more effective treatment options for solid tumors and hematologic malignancies.
Therapeutic vaccines promise new and potentially more effective treatment options for solid tumors and hematologic malignancies.

Vaccines are used to prevent bacterial infections, such as pneumonia, and viral diseases, such as influenza.1 Recently, they are being used to prevent cancers. For example, the human papilloma virus vaccine prevents infection with the virus that is associated with cervical cancer.2 Now there is another purpose for vaccines: They can be used therapeutically. Vaccines can be given even after a person manifests a tumor; they can cause the tumor to shrink or disappear. Therapeutic vaccines hold the potential to effect profound changes in the treatment of several cancers.

Biologics

Biologics, a recently developed class of weapons, can be but aren’t always relatively specific to cancer cells. Biologic agents include inhibitors and monoclonal antibodies. Inhibitors target specific functions, such as angiogenesis. However, inhibitors are not always selective for cancer cells. Angiogenesis inhibitors block formation of blood vessels. Like cytotoxic chemotherapy agents, they block the formation of blood vessels in normal cells as well as cancer cells. Cancer cells, however, generally grow more rapidly than do normal host cells, so they need more blood vessels faster to nourish their cells.3

Some inhibitors are more specific. An inhibitor molecule can be directed against a specific transcript that does not occur in normal cells. In normal cells, the breakpoint cluster region (BCR) gene on chromosome 22 directs synthesis of its protein product, whereas the abelson murine leukemia viral oncogene homolog 1 (ABL1) gene on chromosome 9 specifies another protein. However, in chronic myelogenous leukemia, the ends of those 2 chromosomes translocate, and the BCR/ABL1 transcript is a hybrid of RNA derived from the original chromosome and that of the newly attached, translocated chromosome 9. This hybrid RNA directs synthesis of a fusion protein, a tyrosine kinase; the fusion protein is not produced in normal host cells. Thus imatinib, an inhibitor directed at the fusion protein, inhibits the cancer cells specifically.4

Monoclonal Antibodies

Monoclonal antibodies exploit the host’s immune system to destroy cancer cells. Like the inhibitors, monoclonal antibodies can be directed against a specific functional protein, which is not necessarily specific to tumor cells, or against a protein or family of proteins unique to the cancer cells. For example, trastuzumab is a monoclonal antibody directed against the HER2/neu oncogene, which is amplified in some breast cancers.5 The host’s immune system recognizes the antibodyantigen complex and signals its macrophages to destroy the complex, and the cancer cell dies.

Vaccines also exploit the immune system. One way to immunize against a virus, is to introduce a live or killed virus or a part of a virus—usually a part of a viral protein—into the recipient. The immune system recognizes the foreign virus and makes antibodies. When the immune system is challenged by exposure to the pathogenic virus, the antiviral antibodies recognize and bind to it. The host’s macrophages then engulf the antigenantibody complex and destroy it.6 This type of vaccine prevents infection.

Viral Vaccines

Few vaccines prevent specific cancers. Immunization against the hepatitis B virus, for example, confers immunity to a virus whose infection is a major risk factor for hepatocellular carcinoma.7 Thus it is not truly an anticancer vaccine but rather an antiviral vaccine. Similarly, the HPV
vaccine does not prevent cervical cancer but confers immunity
to a virus that causes cervical cancer.2

Using vaccines to prevent cancer in general is now becoming feasible. Cancer vaccines in use or in development at this time are of 2 general types: those derived from a single tumor and are designed to elicit immunity to that particular tumor in that particular individual host; and those designed to provoke an immune response to that particular type of tumor in any host. The latter type can be mass produced, whereas vaccines of the first type are restricted to the host of origin.

Autologous Vaccines

At the time of biopsy or surgical excision, tumors are mechanically and enzymatically dissociated into single cells, then grown in tissue culture. When they are cultured in suspension with agitation, the cancer stem cells (CSCs) form spheroids, and other cell types do not. The CSCs are the progenitor cells of the specific tumor, much like bone marrow stem cells are the progenitors of the myeloid, erythroid, and megakaryocyte (platelet) cell lineages of human blood. After the CSCs are expanded, the spheroids are harvested, and their RNA is extracted. The messenger RNAs, which specify the proteins made by the CSCs, are converted to complementary DNAs, which are then amplified using the polymerase chain reaction.8

Meanwhile, the patient’s peripheral blood mononuclear cells (PBMCs) are harvested from the blood by leukapheresis. The PBMCs are then enriched for monocytes by immunologic depletion of B and T cells. The monocytes are cultured in the presence of interleukin-4 and granulocytemacrophage-colony-stimulating factor (GM-CSF), and they become immature dendritic cells after 5 days. These immature dendritic cells are transfected with the amplified DNA from the CSC tumor spheres and grown for 2 more days in a medium supplemented with interleukin-1b (IL-1b), interleukin-6 (IL-6), tumor necrosis factor-α, and prostaglandin E2, then tested for markers characteristic of dendritic cells and frozen.

Aliquots of these transfected dendritic cells are injected into the patient at intervals over several months. Dendritic cells, which are the most efficient cells at eliciting an immune response, present the tumor CSC antigens to the patient’s immune system, which develops antibodies to the antigens presented on the dendritic cells and proceeds to destroy the tumor CSCs remaining in the patient’s tumor. Without its CSCs, the tumor is unable to revitalize itself, the rest of the tumor cells eventually die, and the tumor shrinks. The patient is monitored for immune response, and the tumor is regularly imaged. This vaccine is an example of therapeutic use: It can cause tumor regression.8

Sipuleucil-T is another example of an autologous vaccine used for therapeutic purposes. The patient’s antigenpresenting cells (APCs) are harvested by leukopheresis. They are then cultured in the presence of the protein made by fusing prostatic antigen phosphatase (PAP) with the GM-CSF to form the fusion protein PAP-GM-CSF. These modified APCs are then reinjected into the same host. Sipuleucil-T is now FDA approved for the treatment of metastatic castrate-resistant prostate cancer (mCRPC) and has been shown to extend life by approximately 4 months.9

Universal Vaccine for a Specific Tumor

Cancer vaccines can also be prepared in a more conventional manner, analogous to the more commonly used vaccines, and given to all patients with that specific tumor. PROSTVAC is another vaccine for mCRPC as well as for earlier-stage prostate cancer. The DNA specifying prostate-specific antigen (PSA), the secreted protein expressed by the tumor, is mutated very slightly and inserted into a vaccinia virus (Figure). Also inserted into the vaccinia virus are DNAs encoding 3 proteins that
stimulate the patient’s immune system: B7.1, intercellular adhesion molecule-1 (ICAM-1), and leukocyte functionassociated antigen-3 (LFA-3).

This recombinant viral vector is propagated in tissue culture, purified, and administered to the patient subcutaneously. The virus replicates for a short period in the host, expressing the 3 immunostimulatory molecules and the mutated PSA molecule protein product, which differs by 1 amino acid from that expressed by the patient’s tumor. The vaccinia virus and the mutant protein are recognized as foreign by the patient’s immune system, whereas the 3 immunostimulatory molecules are not, as they are the same as the host’s immunostimulatory molecules.

Subsequent boosts, administered over about 5 months, are with a fowlpox virus vector (which does not replicate in the patient) containing the same 4 genes that had been inserted into the vaccinia virus. This strategy enhances the immune response to the mutant tumor product and ensures that the host is not generating a response to the vaccinia virus. The mutant tumor product is sufficiently similar to the patient’s native tumor product that the immune system mounts a response to both proteins. The T cells generated in response to the mutant protein bind to both PSA and mutated PSA molecule proteins, and cells harboring these proteins are destroyed.10

Whereas the T-cell response is important for the activity of the PROSTVAC vaccine, and there is no evidence for formation of anti-PSA antibodies, other vaccines exploit both B- and T-cell responses.10,11 After a virus commandeers the host cell’s replication, transcription, and translation machinery, the macrophage breaks down the viral protein product (the mutated or immunogenic molecule) into small fragments. These fragments bind to major histocompatibility complex (MHC) class II molecules, which are produced by the macrophages. These complexes of antigen (virus) fragments with MHC class II molecules are transported to the surface of the macrophage, where the antigen is presented to lymphocytes. A receptor on a B lymphocyte adheres to the viral antigen and thus becomes an activated B cell, which divides and produces many copies. The B lymphocytes mature into plasma cells and release antibodies that adhere to the virus. A helper T-cell receptor can adhere to the antigen-MHC class II complex displayed on the B cell and activate the lymphocyte, causing the release of more and different cytokines. The cytokines stimulate the activated B cell to divide, producing functionally mature antibodies that recognize and attach to the virus. Macrophages can then engulf and destroy the virus, or the antibody-linked viruses can be excreted in the urine or stool.12

When viruses (eg, the recombinant vaccinia virus of the PROSTVAC vaccine) infect cells and replicate, fragments of viral proteins become attached to MHC class I molecules (see Box). These complexes attach to the cell surface and are presented to cytotoxic T cells. The activated cytotoxic T cells divide and destroy cells harboring the virus or viral antigens.12

Some of the B lymphocytes become memory B cells. Similarly, some of the T lymphocytes become memory T cells. These memory B cells respond and replicate rapidly on re-exposure to the virus or viral proteins to produce antigen-specific antibodies.12

When a macrophage engulfs and breaks down a tumor cell harboring a PSA molecule, other tumor-specific protein fragments are released. These tumor-specific fragments then serve as antigens and elicit an immune response, generating even more antitumor antibodies and T-cell interactions. Thus the tumor regresses. Importantly, this type of vaccine can be mass produced.

Vaccines With Chemotherapy

Another approach to the design of a tumor-specific vaccine is based on tumor-associated peptides (TUMAPs). These TUMAPs are highly overexpressed in tumors relative to a number of normal, healthy tissues. Nine highly immunogenic TUMAPs from 80 specimens of the same tumor type, identified by mass spectrometry, gene expression profiling, literature-based functional assessment, bioinformatics, and human T-cell assays, are used to prepare IMA901, a multipeptide vaccine against the specific tumor, renal cell carcinoma. The pool of selected peptides, composed of 9 to 16 amino acids, is then administered to patients following a dose of the immunomodulator GM-CSF for a total of as many as 17 injections. Low-dose cyclophosphamide administered prior to the first vaccination downregulates regulator T cells, enhancing immune response and resulting in prolonged survival.13 This type of vaccine can also be mass produced.

Conclusions

The vaccines that are currently in use or in clinical trials are being used in patients with advanced disease. As such, they are therapeutic rather than prophylactic. It is inevitable that more vaccines will earn FDA approval. Eventually, they will be used in earlier-stage disease. It is conceivable that some can be used prophylactically when a malignancy first becomes detectable, especially as more sensitive detection methods are developed. Vaccine delivery systems are likely to change as researchers investigate and adopt matrix materials that increase efficacy.

Cancer vaccines hold the potential for greater selectivity in treatment of malignancies. These vaccines may enable us to use cytotoxic chemotherapy agents less often or perhaps more effectively when used in conjunction with vaccines. Vaccines that can be mass produced could conceivably decrease the financial burden of cancer treatment as well as the human cost of malignant diseases and their therapy and care requirements.

Solid-tumor and hematologic malignancy vaccines are coming into wider use as the science is better understood and more methods of generating immune responses are explored. A personalized cancer vaccine, sipuleucel-T, is FDA approved for clinical use against mCRPC.9 A poxvirus-based vaccine, PROSTVAC, also against mCRPC, is in phase 3 clinical trials.10,11 The multipeptide renal cell carcinoma vaccine IMA901 is also in phase 3 clinical trials.13 A personalized vaccine against glioblastoma multiforme CSCs has extended progression-free survival in its initial pretrial study.8,14 Additional glioblastoma, melanoma, and other vaccines are in clinical trials and under development.

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Click here to continue reading.

Vaccines are used to prevent bacterial infections, such as pneumonia, and viral diseases, such as influenza.1 Recently, they are being used to prevent cancers. For example, the human papilloma virus vaccine prevents infection with the virus that is associated with cervical cancer.2 Now there is another purpose for vaccines: They can be used therapeutically. Vaccines can be given even after a person manifests a tumor; they can cause the tumor to shrink or disappear. Therapeutic vaccines hold the potential to effect profound changes in the treatment of several cancers.

Biologics

Biologics, a recently developed class of weapons, can be but aren’t always relatively specific to cancer cells. Biologic agents include inhibitors and monoclonal antibodies. Inhibitors target specific functions, such as angiogenesis. However, inhibitors are not always selective for cancer cells. Angiogenesis inhibitors block formation of blood vessels. Like cytotoxic chemotherapy agents, they block the formation of blood vessels in normal cells as well as cancer cells. Cancer cells, however, generally grow more rapidly than do normal host cells, so they need more blood vessels faster to nourish their cells.3

Some inhibitors are more specific. An inhibitor molecule can be directed against a specific transcript that does not occur in normal cells. In normal cells, the breakpoint cluster region (BCR) gene on chromosome 22 directs synthesis of its protein product, whereas the abelson murine leukemia viral oncogene homolog 1 (ABL1) gene on chromosome 9 specifies another protein. However, in chronic myelogenous leukemia, the ends of those 2 chromosomes translocate, and the BCR/ABL1 transcript is a hybrid of RNA derived from the original chromosome and that of the newly attached, translocated chromosome 9. This hybrid RNA directs synthesis of a fusion protein, a tyrosine kinase; the fusion protein is not produced in normal host cells. Thus imatinib, an inhibitor directed at the fusion protein, inhibits the cancer cells specifically.4

Monoclonal Antibodies

Monoclonal antibodies exploit the host’s immune system to destroy cancer cells. Like the inhibitors, monoclonal antibodies can be directed against a specific functional protein, which is not necessarily specific to tumor cells, or against a protein or family of proteins unique to the cancer cells. For example, trastuzumab is a monoclonal antibody directed against the HER2/neu oncogene, which is amplified in some breast cancers.5 The host’s immune system recognizes the antibodyantigen complex and signals its macrophages to destroy the complex, and the cancer cell dies.

Vaccines also exploit the immune system. One way to immunize against a virus, is to introduce a live or killed virus or a part of a virus—usually a part of a viral protein—into the recipient. The immune system recognizes the foreign virus and makes antibodies. When the immune system is challenged by exposure to the pathogenic virus, the antiviral antibodies recognize and bind to it. The host’s macrophages then engulf the antigenantibody complex and destroy it.6 This type of vaccine prevents infection.

Viral Vaccines

Few vaccines prevent specific cancers. Immunization against the hepatitis B virus, for example, confers immunity to a virus whose infection is a major risk factor for hepatocellular carcinoma.7 Thus it is not truly an anticancer vaccine but rather an antiviral vaccine. Similarly, the HPV
vaccine does not prevent cervical cancer but confers immunity
to a virus that causes cervical cancer.2

Using vaccines to prevent cancer in general is now becoming feasible. Cancer vaccines in use or in development at this time are of 2 general types: those derived from a single tumor and are designed to elicit immunity to that particular tumor in that particular individual host; and those designed to provoke an immune response to that particular type of tumor in any host. The latter type can be mass produced, whereas vaccines of the first type are restricted to the host of origin.

Autologous Vaccines

At the time of biopsy or surgical excision, tumors are mechanically and enzymatically dissociated into single cells, then grown in tissue culture. When they are cultured in suspension with agitation, the cancer stem cells (CSCs) form spheroids, and other cell types do not. The CSCs are the progenitor cells of the specific tumor, much like bone marrow stem cells are the progenitors of the myeloid, erythroid, and megakaryocyte (platelet) cell lineages of human blood. After the CSCs are expanded, the spheroids are harvested, and their RNA is extracted. The messenger RNAs, which specify the proteins made by the CSCs, are converted to complementary DNAs, which are then amplified using the polymerase chain reaction.8

Meanwhile, the patient’s peripheral blood mononuclear cells (PBMCs) are harvested from the blood by leukapheresis. The PBMCs are then enriched for monocytes by immunologic depletion of B and T cells. The monocytes are cultured in the presence of interleukin-4 and granulocytemacrophage-colony-stimulating factor (GM-CSF), and they become immature dendritic cells after 5 days. These immature dendritic cells are transfected with the amplified DNA from the CSC tumor spheres and grown for 2 more days in a medium supplemented with interleukin-1b (IL-1b), interleukin-6 (IL-6), tumor necrosis factor-α, and prostaglandin E2, then tested for markers characteristic of dendritic cells and frozen.

Aliquots of these transfected dendritic cells are injected into the patient at intervals over several months. Dendritic cells, which are the most efficient cells at eliciting an immune response, present the tumor CSC antigens to the patient’s immune system, which develops antibodies to the antigens presented on the dendritic cells and proceeds to destroy the tumor CSCs remaining in the patient’s tumor. Without its CSCs, the tumor is unable to revitalize itself, the rest of the tumor cells eventually die, and the tumor shrinks. The patient is monitored for immune response, and the tumor is regularly imaged. This vaccine is an example of therapeutic use: It can cause tumor regression.8

Sipuleucil-T is another example of an autologous vaccine used for therapeutic purposes. The patient’s antigenpresenting cells (APCs) are harvested by leukopheresis. They are then cultured in the presence of the protein made by fusing prostatic antigen phosphatase (PAP) with the GM-CSF to form the fusion protein PAP-GM-CSF. These modified APCs are then reinjected into the same host. Sipuleucil-T is now FDA approved for the treatment of metastatic castrate-resistant prostate cancer (mCRPC) and has been shown to extend life by approximately 4 months.9

Universal Vaccine for a Specific Tumor

Cancer vaccines can also be prepared in a more conventional manner, analogous to the more commonly used vaccines, and given to all patients with that specific tumor. PROSTVAC is another vaccine for mCRPC as well as for earlier-stage prostate cancer. The DNA specifying prostate-specific antigen (PSA), the secreted protein expressed by the tumor, is mutated very slightly and inserted into a vaccinia virus (Figure). Also inserted into the vaccinia virus are DNAs encoding 3 proteins that
stimulate the patient’s immune system: B7.1, intercellular adhesion molecule-1 (ICAM-1), and leukocyte functionassociated antigen-3 (LFA-3).

This recombinant viral vector is propagated in tissue culture, purified, and administered to the patient subcutaneously. The virus replicates for a short period in the host, expressing the 3 immunostimulatory molecules and the mutated PSA molecule protein product, which differs by 1 amino acid from that expressed by the patient’s tumor. The vaccinia virus and the mutant protein are recognized as foreign by the patient’s immune system, whereas the 3 immunostimulatory molecules are not, as they are the same as the host’s immunostimulatory molecules.

Subsequent boosts, administered over about 5 months, are with a fowlpox virus vector (which does not replicate in the patient) containing the same 4 genes that had been inserted into the vaccinia virus. This strategy enhances the immune response to the mutant tumor product and ensures that the host is not generating a response to the vaccinia virus. The mutant tumor product is sufficiently similar to the patient’s native tumor product that the immune system mounts a response to both proteins. The T cells generated in response to the mutant protein bind to both PSA and mutated PSA molecule proteins, and cells harboring these proteins are destroyed.10

Whereas the T-cell response is important for the activity of the PROSTVAC vaccine, and there is no evidence for formation of anti-PSA antibodies, other vaccines exploit both B- and T-cell responses.10,11 After a virus commandeers the host cell’s replication, transcription, and translation machinery, the macrophage breaks down the viral protein product (the mutated or immunogenic molecule) into small fragments. These fragments bind to major histocompatibility complex (MHC) class II molecules, which are produced by the macrophages. These complexes of antigen (virus) fragments with MHC class II molecules are transported to the surface of the macrophage, where the antigen is presented to lymphocytes. A receptor on a B lymphocyte adheres to the viral antigen and thus becomes an activated B cell, which divides and produces many copies. The B lymphocytes mature into plasma cells and release antibodies that adhere to the virus. A helper T-cell receptor can adhere to the antigen-MHC class II complex displayed on the B cell and activate the lymphocyte, causing the release of more and different cytokines. The cytokines stimulate the activated B cell to divide, producing functionally mature antibodies that recognize and attach to the virus. Macrophages can then engulf and destroy the virus, or the antibody-linked viruses can be excreted in the urine or stool.12

When viruses (eg, the recombinant vaccinia virus of the PROSTVAC vaccine) infect cells and replicate, fragments of viral proteins become attached to MHC class I molecules (see Box). These complexes attach to the cell surface and are presented to cytotoxic T cells. The activated cytotoxic T cells divide and destroy cells harboring the virus or viral antigens.12

Some of the B lymphocytes become memory B cells. Similarly, some of the T lymphocytes become memory T cells. These memory B cells respond and replicate rapidly on re-exposure to the virus or viral proteins to produce antigen-specific antibodies.12

When a macrophage engulfs and breaks down a tumor cell harboring a PSA molecule, other tumor-specific protein fragments are released. These tumor-specific fragments then serve as antigens and elicit an immune response, generating even more antitumor antibodies and T-cell interactions. Thus the tumor regresses. Importantly, this type of vaccine can be mass produced.

Vaccines With Chemotherapy

Another approach to the design of a tumor-specific vaccine is based on tumor-associated peptides (TUMAPs). These TUMAPs are highly overexpressed in tumors relative to a number of normal, healthy tissues. Nine highly immunogenic TUMAPs from 80 specimens of the same tumor type, identified by mass spectrometry, gene expression profiling, literature-based functional assessment, bioinformatics, and human T-cell assays, are used to prepare IMA901, a multipeptide vaccine against the specific tumor, renal cell carcinoma. The pool of selected peptides, composed of 9 to 16 amino acids, is then administered to patients following a dose of the immunomodulator GM-CSF for a total of as many as 17 injections. Low-dose cyclophosphamide administered prior to the first vaccination downregulates regulator T cells, enhancing immune response and resulting in prolonged survival.13 This type of vaccine can also be mass produced.

Conclusions

The vaccines that are currently in use or in clinical trials are being used in patients with advanced disease. As such, they are therapeutic rather than prophylactic. It is inevitable that more vaccines will earn FDA approval. Eventually, they will be used in earlier-stage disease. It is conceivable that some can be used prophylactically when a malignancy first becomes detectable, especially as more sensitive detection methods are developed. Vaccine delivery systems are likely to change as researchers investigate and adopt matrix materials that increase efficacy.

Cancer vaccines hold the potential for greater selectivity in treatment of malignancies. These vaccines may enable us to use cytotoxic chemotherapy agents less often or perhaps more effectively when used in conjunction with vaccines. Vaccines that can be mass produced could conceivably decrease the financial burden of cancer treatment as well as the human cost of malignant diseases and their therapy and care requirements.

Solid-tumor and hematologic malignancy vaccines are coming into wider use as the science is better understood and more methods of generating immune responses are explored. A personalized cancer vaccine, sipuleucel-T, is FDA approved for clinical use against mCRPC.9 A poxvirus-based vaccine, PROSTVAC, also against mCRPC, is in phase 3 clinical trials.10,11 The multipeptide renal cell carcinoma vaccine IMA901 is also in phase 3 clinical trials.13 A personalized vaccine against glioblastoma multiforme CSCs has extended progression-free survival in its initial pretrial study.8,14 Additional glioblastoma, melanoma, and other vaccines are in clinical trials and under development.

Author disclosures
The author reports no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the author and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Click here to continue reading.

References

1. U.S. Department of Health and Human Services, National Institutes of Health, National Institute of Allergy and Infectious Diseases. Understanding vaccines: what they are, how they work. National Institute of Allergy and Infectious Diseases Website. http://www.niaid.nih.gov/topics/vaccines/documents/undvacc.pdf. Published January 2008. Accessed March 24, 2015. .

2. Hagensee ME, Yaegashi N, Galloway GA. Self-assembly of human papilloma virus type 1 capsids by expression of the L1 protein alone or by coexpression of the L1 and L2 capsid proteins. J Virol. 1993;67(1):315-322.

3. American Cancer Society. Anti-angiogenesis treatment. American Cancer Society Website. http://www.cancer.org/acs/groups/cid/documents/webcontent/002988-pdf.pdf. Revised March 10, 2009. Accessed March 24, 2015.

4. Gambacorti-Passerini CB, Gunby RH, Piazza R, Galietta A, Rostagno R, Scapozza L. Molecular mechanisms of resistance to imatinib in Philadelphia-chromosomepositive leukaemias. Lancet Oncol. 2003;4(2):75-85.

5. Hudis CA. Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39-51.

6. NPI reference guide on vaccines and vaccine safety: How vaccines work. PATH Website. http://www.path.org/vaccineresources/files/How_Vaccines_Work.pdf. Accessed March 24, 2015.

7. Beasley RP, Hwang LY, Lin CC, Chien CS. Hepatocellular carcinoma and hepatitis B virus: a prospective study of 22 707 men in Taiwan. Lancet. 1981;318(8256):1129-1133.

8. Vik-Mo EO, Nyakas M, Mikkelsen BV, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62(9):1499-1509.

9. Kantoff PW, Higano CS, Shore ND, et al; IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411-422.

10. Gulley JL, Madan RA, Tsang KY, et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res. 2014;2(2):133-141.

11. Campbell CT, Gulley JL, Oyelaran O, Hodge JW, Schlom J, Gildersleeve JC. Humoral response to a viral glycan correlates with survival on PROSTVAC VF. Proc Natl Acad Sci USA. 2014;111(17):E1749-E1758.

12. Public Health England. Immunity and how vaccines work: The green book, chapter 1. GOV.UK Website. https://www.gov.uk/government/publications/immunity-and-how-vaccines-work-the-green-book-chapter-1. Published March 19, 2013. Accessed March 24, 2015.

13. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18(8):1254-1261.

14. Ottenhausen M, Bodhinayake I, Banu M, Kesavabhotla K, Ray A, Boockvar JA. Industry progress report on neuro-oncology: biotech update 2013. J Neurooncol. 2013;115(2):311-316.

References

1. U.S. Department of Health and Human Services, National Institutes of Health, National Institute of Allergy and Infectious Diseases. Understanding vaccines: what they are, how they work. National Institute of Allergy and Infectious Diseases Website. http://www.niaid.nih.gov/topics/vaccines/documents/undvacc.pdf. Published January 2008. Accessed March 24, 2015. .

2. Hagensee ME, Yaegashi N, Galloway GA. Self-assembly of human papilloma virus type 1 capsids by expression of the L1 protein alone or by coexpression of the L1 and L2 capsid proteins. J Virol. 1993;67(1):315-322.

3. American Cancer Society. Anti-angiogenesis treatment. American Cancer Society Website. http://www.cancer.org/acs/groups/cid/documents/webcontent/002988-pdf.pdf. Revised March 10, 2009. Accessed March 24, 2015.

4. Gambacorti-Passerini CB, Gunby RH, Piazza R, Galietta A, Rostagno R, Scapozza L. Molecular mechanisms of resistance to imatinib in Philadelphia-chromosomepositive leukaemias. Lancet Oncol. 2003;4(2):75-85.

5. Hudis CA. Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39-51.

6. NPI reference guide on vaccines and vaccine safety: How vaccines work. PATH Website. http://www.path.org/vaccineresources/files/How_Vaccines_Work.pdf. Accessed March 24, 2015.

7. Beasley RP, Hwang LY, Lin CC, Chien CS. Hepatocellular carcinoma and hepatitis B virus: a prospective study of 22 707 men in Taiwan. Lancet. 1981;318(8256):1129-1133.

8. Vik-Mo EO, Nyakas M, Mikkelsen BV, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62(9):1499-1509.

9. Kantoff PW, Higano CS, Shore ND, et al; IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411-422.

10. Gulley JL, Madan RA, Tsang KY, et al. Immune impact induced by PROSTVAC (PSA-TRICOM), a therapeutic vaccine for prostate cancer. Cancer Immunol Res. 2014;2(2):133-141.

11. Campbell CT, Gulley JL, Oyelaran O, Hodge JW, Schlom J, Gildersleeve JC. Humoral response to a viral glycan correlates with survival on PROSTVAC VF. Proc Natl Acad Sci USA. 2014;111(17):E1749-E1758.

12. Public Health England. Immunity and how vaccines work: The green book, chapter 1. GOV.UK Website. https://www.gov.uk/government/publications/immunity-and-how-vaccines-work-the-green-book-chapter-1. Published March 19, 2013. Accessed March 24, 2015.

13. Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med. 2012;18(8):1254-1261.

14. Ottenhausen M, Bodhinayake I, Banu M, Kesavabhotla K, Ray A, Boockvar JA. Industry progress report on neuro-oncology: biotech update 2013. J Neurooncol. 2013;115(2):311-316.

Issue
Federal Practitioner - 32(4)s
Issue
Federal Practitioner - 32(4)s
Page Number
27S-30S
Page Number
27S-30S
Publications
Publications
Topics
Article Type
Display Headline
Harnessing Vaccines to Treat Cancers
Display Headline
Harnessing Vaccines to Treat Cancers
Legacy Keywords
cancer vaccines, biologics, biologic agents, angiogenesis inhibitors, monoclonal antibodies, viral vaccines, autologous vaccines, poxvirus vector, PROSTVAC, vaccines with chemotherapy, tumor-specific vaccine, tumor-associated peptides, TUMAPs, sipuleucel-T personalized cancer vaccine
Legacy Keywords
cancer vaccines, biologics, biologic agents, angiogenesis inhibitors, monoclonal antibodies, viral vaccines, autologous vaccines, poxvirus vector, PROSTVAC, vaccines with chemotherapy, tumor-specific vaccine, tumor-associated peptides, TUMAPs, sipuleucel-T personalized cancer vaccine
Sections
Citation Override
Fed Pract. 2015 May;32(suppl 4):27S-30S
Disallow All Ads
Alternative CME

NRAS mutations predict immunotherapy outcomes in melanoma patients

Article Type
Changed
Mon, 01/14/2019 - 09:11
Display Headline
NRAS mutations predict immunotherapy outcomes in melanoma patients

Patients with advanced melanoma who were treated with immunotherapy responded better if they harbored mutations in the NRAS gene, according to a study published March 3 in Cancer Immunology Research.

Out of 229 cases retrospectively analyzed, 26% had mutations in NRASG12/G13/Q61, 23% had BRAFV600, and 51% were wild type for NRAS and BRAF. Patients received first-line therapy with high-dose IL-2 (25%), ipilimumab (62%), or anti-PD-1/PD-L1 (12%), investigators reported (Cancer Immunol. Res. 2015 March 3).

 

©benjaminalbiach/ThinkStock

Complete or partial responses were found in 32% of patients with NRAS-mutant melanomas, compared with 20% of those without NRAS mutations (P = .07). Clinical benefit (defined as complete response, partial response, or stable disease for 24 weeks or longer) was observed in 50% of the NRAS mutant group vs. 30% of the non–mutant NRAS group (P < .01), reported Dr. Douglas B. Johnson of Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, Tenn., and associates.

Although the numbers for individual agents were small, the NRAS-mutant benefit was most pronounced for immune checkpoint inhibitors, especially anti-PD-1/PD-L1 therapy, where clinical benefit was observed in 8 of 11 NRAS-mutant patients vs. 13 of 37 patients with wild-type NRAS.

“This finding could have implications for molecular testing and treatment decision making, and it provides early insights into the complex relationship between tumor genetics and the immune response,” Dr. Johnson and associates wrote.

Patients with NRAS-mutant melanoma account for 15%-20% of all melanomas, and the mutation is associated with a poorer prognosis. The authors speculate that elevated PD-1 expression may be a factor in inferior prognosis of NRAS-mutant phenotypes as well as the observed improved response to anti-PD-1.

“We studied a small group of patients, but the results were quite suggestive. Our findings need to be confirmed in a prospective study. This study highlights the need to find predictive markers that can help us understand which patients will respond to therapy. Our study will hopefully lead to understanding the biological mechanisms that explain why NRAS mutations predict response,” Dr. Johnson and his associates said.

Click for Credit Link
Publications
Topics
Legacy Keywords
melanoma, NRAS, immunotherapy
Click for Credit Link
Click for Credit Link

Patients with advanced melanoma who were treated with immunotherapy responded better if they harbored mutations in the NRAS gene, according to a study published March 3 in Cancer Immunology Research.

Out of 229 cases retrospectively analyzed, 26% had mutations in NRASG12/G13/Q61, 23% had BRAFV600, and 51% were wild type for NRAS and BRAF. Patients received first-line therapy with high-dose IL-2 (25%), ipilimumab (62%), or anti-PD-1/PD-L1 (12%), investigators reported (Cancer Immunol. Res. 2015 March 3).

 

©benjaminalbiach/ThinkStock

Complete or partial responses were found in 32% of patients with NRAS-mutant melanomas, compared with 20% of those without NRAS mutations (P = .07). Clinical benefit (defined as complete response, partial response, or stable disease for 24 weeks or longer) was observed in 50% of the NRAS mutant group vs. 30% of the non–mutant NRAS group (P < .01), reported Dr. Douglas B. Johnson of Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, Tenn., and associates.

Although the numbers for individual agents were small, the NRAS-mutant benefit was most pronounced for immune checkpoint inhibitors, especially anti-PD-1/PD-L1 therapy, where clinical benefit was observed in 8 of 11 NRAS-mutant patients vs. 13 of 37 patients with wild-type NRAS.

“This finding could have implications for molecular testing and treatment decision making, and it provides early insights into the complex relationship between tumor genetics and the immune response,” Dr. Johnson and associates wrote.

Patients with NRAS-mutant melanoma account for 15%-20% of all melanomas, and the mutation is associated with a poorer prognosis. The authors speculate that elevated PD-1 expression may be a factor in inferior prognosis of NRAS-mutant phenotypes as well as the observed improved response to anti-PD-1.

“We studied a small group of patients, but the results were quite suggestive. Our findings need to be confirmed in a prospective study. This study highlights the need to find predictive markers that can help us understand which patients will respond to therapy. Our study will hopefully lead to understanding the biological mechanisms that explain why NRAS mutations predict response,” Dr. Johnson and his associates said.

Patients with advanced melanoma who were treated with immunotherapy responded better if they harbored mutations in the NRAS gene, according to a study published March 3 in Cancer Immunology Research.

Out of 229 cases retrospectively analyzed, 26% had mutations in NRASG12/G13/Q61, 23% had BRAFV600, and 51% were wild type for NRAS and BRAF. Patients received first-line therapy with high-dose IL-2 (25%), ipilimumab (62%), or anti-PD-1/PD-L1 (12%), investigators reported (Cancer Immunol. Res. 2015 March 3).

 

©benjaminalbiach/ThinkStock

Complete or partial responses were found in 32% of patients with NRAS-mutant melanomas, compared with 20% of those without NRAS mutations (P = .07). Clinical benefit (defined as complete response, partial response, or stable disease for 24 weeks or longer) was observed in 50% of the NRAS mutant group vs. 30% of the non–mutant NRAS group (P < .01), reported Dr. Douglas B. Johnson of Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, Tenn., and associates.

Although the numbers for individual agents were small, the NRAS-mutant benefit was most pronounced for immune checkpoint inhibitors, especially anti-PD-1/PD-L1 therapy, where clinical benefit was observed in 8 of 11 NRAS-mutant patients vs. 13 of 37 patients with wild-type NRAS.

“This finding could have implications for molecular testing and treatment decision making, and it provides early insights into the complex relationship between tumor genetics and the immune response,” Dr. Johnson and associates wrote.

Patients with NRAS-mutant melanoma account for 15%-20% of all melanomas, and the mutation is associated with a poorer prognosis. The authors speculate that elevated PD-1 expression may be a factor in inferior prognosis of NRAS-mutant phenotypes as well as the observed improved response to anti-PD-1.

“We studied a small group of patients, but the results were quite suggestive. Our findings need to be confirmed in a prospective study. This study highlights the need to find predictive markers that can help us understand which patients will respond to therapy. Our study will hopefully lead to understanding the biological mechanisms that explain why NRAS mutations predict response,” Dr. Johnson and his associates said.

Publications
Publications
Topics
Article Type
Display Headline
NRAS mutations predict immunotherapy outcomes in melanoma patients
Display Headline
NRAS mutations predict immunotherapy outcomes in melanoma patients
Legacy Keywords
melanoma, NRAS, immunotherapy
Legacy Keywords
melanoma, NRAS, immunotherapy
Article Source

FROM CANCER IMMUNOLOGY RESEARCH

Disallow All Ads
Vitals

Key clinical point: Patients with advanced melanoma and mutations in the NRAS gene had better responses to immunotherapy than did those without NRAS mutations.

Major finding: Of the patients with mutated NRAS, 28% had complete or partial responses to first-line immunotherapy, compared with 16% of patients without NRAS mutations.

Data source: The retrospective study evaluated medical records from 229 patients with advanced melanoma who were treated with ipilimumab, IL-2, or anti-PD-1/PD-L1.

Disclosures: Dr. Johnson had no disclosures. Dr. Lovly received grants from AstraZeneca and Novartis. Dr. Iafrate has ownership in ArcherDx and an advisory role with BioReference Labs.