Clinical Edge Journal Scan Commentary: HCC November 2021

Article Type
Changed
Thu, 05/12/2022 - 11:56
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD
Many patients with hepatocellular carcinoma (HCC) require systemic therapy at some point in their treatment course. This month, we will review articles that analyze outcomes of systemic treatments, either on their own, or in combination with liver-directed therapy.

Tyrosine kinase inhibitors are the mainstay of systemic HCC therapy, however treatment at FDA-approved doses frequently leads to unacceptable toxicities, leading to reductions in the prescribed dose. Tokunaga et al. investigated whether lenvatinib dose intensity affects outcomes of patients with unresectable HCC. This was a retrospective analysis of 100 patients who received lenvatinib in the first- or later-line settings. Fifty-one patients started lenvatinib at the standard dose and 49 patients at a reduced dose. Dose reduction was carried out in 29 patients during cycle 1, and 62 patients during all cycles, with the cumulative dose reduction rate in all cycles of 79.9%.  Upon analysis, the authors confirmed that tumor responses and stable disease on lenvatinib correlated favorably with overall survival (OS) and time to progression (TTP). In addition, they found that higher dose intensity correlated with a higher response rate, though most (56%) patients were unable to maintain the recommended dose intensity due to unacceptable adverse events. In the final analysis, dose modification was not negatively associated with OS, TTP, or disease control with lenvatinib. Therefore, it remains reasonable to adjust the dose of lenvatinib to minimize toxicity that would affect adversely patient quality of life. Disease control remains the best predictor of longer survival, though it does not seem that highest doses of lenvatinib are needed to achieve that benefit.

Cabozantinib is approved for previously treated patients with unresectable HCC based on the phase III CELESTIAL trial that enrolled patients with Child Pugh A liver disease who had received up to two previous systemic treatments, one of which was sorafenib. This study demonstrated a statistically significant improvement in overall- and disease-free survival. Kelley et al analyzed the outcomes based on the albumin-bilirubin (ALBI) grade, an objective measure of liver function, of patients in the CELESTIAL trial. ALBI scores were retrospectively calculated based on baseline serum albumin and total bilirubin. The median OS was 17.5 months in the cabozantinib arm versus 11.4 months in the placebo arm for the ALBI grade 1 subgroup, and 8.0 months in the cabozantinib arm versus 6.4 months in the placebo arm for the ALBI grade 2 subgroup. The authors concluded that cabozantinib benefits patients with unresectable HCC irrespective of their ALBI grade, though liver dysfunction remains a poor prognostic indicator in patients with HCC.

Finally, Liu et al analyzed 27 patients with HCC (8 with extrahepatic spread) who received a combination of chemotherapy via hepatic artery infusion, anti-PD-1 immunotherapy, and tyrosine kinase inhibitor. Combination chemotherapy and immunotherapy resulted in a median progression-free survival of 10.6 months with a median 12.9 months’ follow up; the objective response rate was 63.0% and the disease control rate was 92.6%. The authors concluded that this combination of therapies was effective and well-tolerated, with a confirmatory phase 3 study planned.

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology,  Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Publications
Topics
Sections
Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology,  Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology,  Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Dr. Damjanov scans the journals, so you don’t have to!
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD
Many patients with hepatocellular carcinoma (HCC) require systemic therapy at some point in their treatment course. This month, we will review articles that analyze outcomes of systemic treatments, either on their own, or in combination with liver-directed therapy.

Tyrosine kinase inhibitors are the mainstay of systemic HCC therapy, however treatment at FDA-approved doses frequently leads to unacceptable toxicities, leading to reductions in the prescribed dose. Tokunaga et al. investigated whether lenvatinib dose intensity affects outcomes of patients with unresectable HCC. This was a retrospective analysis of 100 patients who received lenvatinib in the first- or later-line settings. Fifty-one patients started lenvatinib at the standard dose and 49 patients at a reduced dose. Dose reduction was carried out in 29 patients during cycle 1, and 62 patients during all cycles, with the cumulative dose reduction rate in all cycles of 79.9%.  Upon analysis, the authors confirmed that tumor responses and stable disease on lenvatinib correlated favorably with overall survival (OS) and time to progression (TTP). In addition, they found that higher dose intensity correlated with a higher response rate, though most (56%) patients were unable to maintain the recommended dose intensity due to unacceptable adverse events. In the final analysis, dose modification was not negatively associated with OS, TTP, or disease control with lenvatinib. Therefore, it remains reasonable to adjust the dose of lenvatinib to minimize toxicity that would affect adversely patient quality of life. Disease control remains the best predictor of longer survival, though it does not seem that highest doses of lenvatinib are needed to achieve that benefit.

Cabozantinib is approved for previously treated patients with unresectable HCC based on the phase III CELESTIAL trial that enrolled patients with Child Pugh A liver disease who had received up to two previous systemic treatments, one of which was sorafenib. This study demonstrated a statistically significant improvement in overall- and disease-free survival. Kelley et al analyzed the outcomes based on the albumin-bilirubin (ALBI) grade, an objective measure of liver function, of patients in the CELESTIAL trial. ALBI scores were retrospectively calculated based on baseline serum albumin and total bilirubin. The median OS was 17.5 months in the cabozantinib arm versus 11.4 months in the placebo arm for the ALBI grade 1 subgroup, and 8.0 months in the cabozantinib arm versus 6.4 months in the placebo arm for the ALBI grade 2 subgroup. The authors concluded that cabozantinib benefits patients with unresectable HCC irrespective of their ALBI grade, though liver dysfunction remains a poor prognostic indicator in patients with HCC.

Finally, Liu et al analyzed 27 patients with HCC (8 with extrahepatic spread) who received a combination of chemotherapy via hepatic artery infusion, anti-PD-1 immunotherapy, and tyrosine kinase inhibitor. Combination chemotherapy and immunotherapy resulted in a median progression-free survival of 10.6 months with a median 12.9 months’ follow up; the objective response rate was 63.0% and the disease control rate was 92.6%. The authors concluded that this combination of therapies was effective and well-tolerated, with a confirmatory phase 3 study planned.

Nevena Damjanov, MD
Many patients with hepatocellular carcinoma (HCC) require systemic therapy at some point in their treatment course. This month, we will review articles that analyze outcomes of systemic treatments, either on their own, or in combination with liver-directed therapy.

Tyrosine kinase inhibitors are the mainstay of systemic HCC therapy, however treatment at FDA-approved doses frequently leads to unacceptable toxicities, leading to reductions in the prescribed dose. Tokunaga et al. investigated whether lenvatinib dose intensity affects outcomes of patients with unresectable HCC. This was a retrospective analysis of 100 patients who received lenvatinib in the first- or later-line settings. Fifty-one patients started lenvatinib at the standard dose and 49 patients at a reduced dose. Dose reduction was carried out in 29 patients during cycle 1, and 62 patients during all cycles, with the cumulative dose reduction rate in all cycles of 79.9%.  Upon analysis, the authors confirmed that tumor responses and stable disease on lenvatinib correlated favorably with overall survival (OS) and time to progression (TTP). In addition, they found that higher dose intensity correlated with a higher response rate, though most (56%) patients were unable to maintain the recommended dose intensity due to unacceptable adverse events. In the final analysis, dose modification was not negatively associated with OS, TTP, or disease control with lenvatinib. Therefore, it remains reasonable to adjust the dose of lenvatinib to minimize toxicity that would affect adversely patient quality of life. Disease control remains the best predictor of longer survival, though it does not seem that highest doses of lenvatinib are needed to achieve that benefit.

Cabozantinib is approved for previously treated patients with unresectable HCC based on the phase III CELESTIAL trial that enrolled patients with Child Pugh A liver disease who had received up to two previous systemic treatments, one of which was sorafenib. This study demonstrated a statistically significant improvement in overall- and disease-free survival. Kelley et al analyzed the outcomes based on the albumin-bilirubin (ALBI) grade, an objective measure of liver function, of patients in the CELESTIAL trial. ALBI scores were retrospectively calculated based on baseline serum albumin and total bilirubin. The median OS was 17.5 months in the cabozantinib arm versus 11.4 months in the placebo arm for the ALBI grade 1 subgroup, and 8.0 months in the cabozantinib arm versus 6.4 months in the placebo arm for the ALBI grade 2 subgroup. The authors concluded that cabozantinib benefits patients with unresectable HCC irrespective of their ALBI grade, though liver dysfunction remains a poor prognostic indicator in patients with HCC.

Finally, Liu et al analyzed 27 patients with HCC (8 with extrahepatic spread) who received a combination of chemotherapy via hepatic artery infusion, anti-PD-1 immunotherapy, and tyrosine kinase inhibitor. Combination chemotherapy and immunotherapy resulted in a median progression-free survival of 10.6 months with a median 12.9 months’ follow up; the objective response rate was 63.0% and the disease control rate was 92.6%. The authors concluded that this combination of therapies was effective and well-tolerated, with a confirmatory phase 3 study planned.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: HCC November 2021
Gate On Date
Mon, 05/17/2021 - 15:00
Un-Gate On Date
Mon, 05/17/2021 - 15:00
Use ProPublica
CFC Schedule Remove Status
Mon, 05/17/2021 - 15:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: CRC November 2021

Article Type
Changed
Fri, 05/06/2022 - 16:51
Dr. Weinberg scans the journals, so you don’t have to!

Benjamin A. Weinberg, MD
We are constantly on the hunt for new, effective therapies for patients with metastatic colorectal cancer (mCRC), the second-leading cause of cancer-related death in the United States after lung cancer. Approximately half of these tumors are driven by mutations in the RAS family of oncogenes which have been historically thought to be undruggable despite extensive research on this front, including the National Cancer Institute’s RAS Initiative. Colorectal cancers also frequently have mutations in TP53, a tumor suppressor gene that is the most commonly mutated gene across all cancers and also has been very difficult to target therapeutically.

The tide may finally be shifting as drugs targeting specific KRAS mutations have made their way into the clinic, and there is particular excitement around the oral WEE1 inhibitor, adavosertib (AZD1775). WEE1 is a cell cycle regulatory protein and WEE1 inhibition may have increased activity in tumors with DNA repair deficiency. In a phase 2 maintenance study, Seligmann and colleagues randomized 69 patients with RAS- and TP53-mutated mCRC with stable disease or better after 16 weeks of induction chemotherapy 2:1 to receive adavosertib vs active monitoring. Median progression-free survival (mPFS) was significant improved with adavosertib (3.61 vs 1.68 months), and patients with left-sided primary tumors appears to derive more benefit. While this finding needs to be further explored in larger clinical trials, it is exciting that there may finally be a new treatment option for patients with this specific molecular subtype of mCRC.

Another maintenance mCRC trial of interest is the PANAMA trial, a phase 2 study in which 248 patients with RAS wild-type mCRC were randomized 1:1 to 5-fluorouracil/leucovorin with or without panitumumab, an anti-epidermal growth factor receptor antibody, following induction chemotherapy with 6 cycles FOLFOX/panitumumab. Modest and co-workers report that mPFS was significantly improved with continuing panitumumab in the maintenance setting (8.8 vs. 5.7 months), and there was a trend towards an overall survival benefit as well. This study further supports continuing anti-EGFR therapy with maintenance chemotherapy for patients with RAS wild-type mCRC.

Finally, in stage III CRC, there is a big movement towards using circulating tumor DNA (ctDNA) as a method to monitor disease recurrence by detecting minimal residual disease based on tumor DNA being shed into the bloodstream. Henriksen et al. evaluated 168 patients with stage III CRC who underwent surgical resection and plasma ctDNA testing using 16 patient-specific DNA variants (tumor tissue-informed testing). The rates of recurrence were much higher in patients with detectable ctDNA post-operatively and/or after the completion of adjuvant chemotherapy, whereas those patients with persistently undetectable ctDNA did not recur. ctDNA is a powerful new technology that we are still learning how to best harness in the clinic, and this study demonstrates its prognostic value and potential ability to detect recurrence prior to standard imaging surveillance. Moreover, the rate of ctDNA rise was also prognostic of survival. ctDNA testing is likely to become standard of care in the management of stage II/III colorectal cancer in the very near future, and we hope that eventually it may be able to predict who needs to receive adjuvant chemotherapy and who does not.

Author and Disclosure Information

Benjamin A. Weinberg, MD

Disclosures: Speaker/advisor for Bayer, Taiho, Sirtex, HalioDx, Lilly, AstraZeneca/Daiichi Sankyo, research support from Ipsen, G1 Therapeutics.

Publications
Topics
Sections
Author and Disclosure Information

Benjamin A. Weinberg, MD

Disclosures: Speaker/advisor for Bayer, Taiho, Sirtex, HalioDx, Lilly, AstraZeneca/Daiichi Sankyo, research support from Ipsen, G1 Therapeutics.

Author and Disclosure Information

Benjamin A. Weinberg, MD

Disclosures: Speaker/advisor for Bayer, Taiho, Sirtex, HalioDx, Lilly, AstraZeneca/Daiichi Sankyo, research support from Ipsen, G1 Therapeutics.

Dr. Weinberg scans the journals, so you don’t have to!
Dr. Weinberg scans the journals, so you don’t have to!

Benjamin A. Weinberg, MD
We are constantly on the hunt for new, effective therapies for patients with metastatic colorectal cancer (mCRC), the second-leading cause of cancer-related death in the United States after lung cancer. Approximately half of these tumors are driven by mutations in the RAS family of oncogenes which have been historically thought to be undruggable despite extensive research on this front, including the National Cancer Institute’s RAS Initiative. Colorectal cancers also frequently have mutations in TP53, a tumor suppressor gene that is the most commonly mutated gene across all cancers and also has been very difficult to target therapeutically.

The tide may finally be shifting as drugs targeting specific KRAS mutations have made their way into the clinic, and there is particular excitement around the oral WEE1 inhibitor, adavosertib (AZD1775). WEE1 is a cell cycle regulatory protein and WEE1 inhibition may have increased activity in tumors with DNA repair deficiency. In a phase 2 maintenance study, Seligmann and colleagues randomized 69 patients with RAS- and TP53-mutated mCRC with stable disease or better after 16 weeks of induction chemotherapy 2:1 to receive adavosertib vs active monitoring. Median progression-free survival (mPFS) was significant improved with adavosertib (3.61 vs 1.68 months), and patients with left-sided primary tumors appears to derive more benefit. While this finding needs to be further explored in larger clinical trials, it is exciting that there may finally be a new treatment option for patients with this specific molecular subtype of mCRC.

Another maintenance mCRC trial of interest is the PANAMA trial, a phase 2 study in which 248 patients with RAS wild-type mCRC were randomized 1:1 to 5-fluorouracil/leucovorin with or without panitumumab, an anti-epidermal growth factor receptor antibody, following induction chemotherapy with 6 cycles FOLFOX/panitumumab. Modest and co-workers report that mPFS was significantly improved with continuing panitumumab in the maintenance setting (8.8 vs. 5.7 months), and there was a trend towards an overall survival benefit as well. This study further supports continuing anti-EGFR therapy with maintenance chemotherapy for patients with RAS wild-type mCRC.

Finally, in stage III CRC, there is a big movement towards using circulating tumor DNA (ctDNA) as a method to monitor disease recurrence by detecting minimal residual disease based on tumor DNA being shed into the bloodstream. Henriksen et al. evaluated 168 patients with stage III CRC who underwent surgical resection and plasma ctDNA testing using 16 patient-specific DNA variants (tumor tissue-informed testing). The rates of recurrence were much higher in patients with detectable ctDNA post-operatively and/or after the completion of adjuvant chemotherapy, whereas those patients with persistently undetectable ctDNA did not recur. ctDNA is a powerful new technology that we are still learning how to best harness in the clinic, and this study demonstrates its prognostic value and potential ability to detect recurrence prior to standard imaging surveillance. Moreover, the rate of ctDNA rise was also prognostic of survival. ctDNA testing is likely to become standard of care in the management of stage II/III colorectal cancer in the very near future, and we hope that eventually it may be able to predict who needs to receive adjuvant chemotherapy and who does not.

Benjamin A. Weinberg, MD
We are constantly on the hunt for new, effective therapies for patients with metastatic colorectal cancer (mCRC), the second-leading cause of cancer-related death in the United States after lung cancer. Approximately half of these tumors are driven by mutations in the RAS family of oncogenes which have been historically thought to be undruggable despite extensive research on this front, including the National Cancer Institute’s RAS Initiative. Colorectal cancers also frequently have mutations in TP53, a tumor suppressor gene that is the most commonly mutated gene across all cancers and also has been very difficult to target therapeutically.

The tide may finally be shifting as drugs targeting specific KRAS mutations have made their way into the clinic, and there is particular excitement around the oral WEE1 inhibitor, adavosertib (AZD1775). WEE1 is a cell cycle regulatory protein and WEE1 inhibition may have increased activity in tumors with DNA repair deficiency. In a phase 2 maintenance study, Seligmann and colleagues randomized 69 patients with RAS- and TP53-mutated mCRC with stable disease or better after 16 weeks of induction chemotherapy 2:1 to receive adavosertib vs active monitoring. Median progression-free survival (mPFS) was significant improved with adavosertib (3.61 vs 1.68 months), and patients with left-sided primary tumors appears to derive more benefit. While this finding needs to be further explored in larger clinical trials, it is exciting that there may finally be a new treatment option for patients with this specific molecular subtype of mCRC.

Another maintenance mCRC trial of interest is the PANAMA trial, a phase 2 study in which 248 patients with RAS wild-type mCRC were randomized 1:1 to 5-fluorouracil/leucovorin with or without panitumumab, an anti-epidermal growth factor receptor antibody, following induction chemotherapy with 6 cycles FOLFOX/panitumumab. Modest and co-workers report that mPFS was significantly improved with continuing panitumumab in the maintenance setting (8.8 vs. 5.7 months), and there was a trend towards an overall survival benefit as well. This study further supports continuing anti-EGFR therapy with maintenance chemotherapy for patients with RAS wild-type mCRC.

Finally, in stage III CRC, there is a big movement towards using circulating tumor DNA (ctDNA) as a method to monitor disease recurrence by detecting minimal residual disease based on tumor DNA being shed into the bloodstream. Henriksen et al. evaluated 168 patients with stage III CRC who underwent surgical resection and plasma ctDNA testing using 16 patient-specific DNA variants (tumor tissue-informed testing). The rates of recurrence were much higher in patients with detectable ctDNA post-operatively and/or after the completion of adjuvant chemotherapy, whereas those patients with persistently undetectable ctDNA did not recur. ctDNA is a powerful new technology that we are still learning how to best harness in the clinic, and this study demonstrates its prognostic value and potential ability to detect recurrence prior to standard imaging surveillance. Moreover, the rate of ctDNA rise was also prognostic of survival. ctDNA testing is likely to become standard of care in the management of stage II/III colorectal cancer in the very near future, and we hope that eventually it may be able to predict who needs to receive adjuvant chemotherapy and who does not.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: CRC November 2021
Gate On Date
Fri, 08/27/2021 - 19:30
Un-Gate On Date
Fri, 08/27/2021 - 19:30
Use ProPublica
CFC Schedule Remove Status
Fri, 08/27/2021 - 19:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: CRC October 2021

Article Type
Changed
Fri, 05/06/2022 - 16:50
Dr. Weinberg scans the journals, so you don’t have to!

Benjamin A. Weinberg, MD, FACP
The choice of therapy in patients with refractory metastatic colorectal cancer (mCRC) can be perplexing. Following first- and second-line chemotherapy regimens including fluoropyrimidines, oxaliplatin, irinotecan, and biologics such as bevacizumab (an anti-vascular endothelial growth factor [anti-VEGF] antibody) or cetuximab (an anti-epidermal growth factor receptor [anti-EGFR] antibody), standard later-line options include regorafenib, trifluridine/tipiracil, or rechallenging with chemotherapy and/or an anti-EGFR antibody.

Emerging data have helped to elucidate the real world experience of regorafenib vs trifluridine/tipiracil in the third-line setting. In a single-institution retrospective study, Patel and colleagues evaluated 126 patients who received trifluridine/tipiracil and 95 patients who received regorafenib. There were higher response and disease control rates seen with trifluridine/tipiracil, although median overall survival was similar (7.5 vs 7.1 months). These findings are limited by the single-institution, retrospective and non-randomized nature of the study. Also, the better-tolerated ReDOS dose-escalation schema for regorafenib was not utilized in this study, and it would be interesting to see how this novel dosing strategy compares to trifluridine/tipiracil in the real world setting.

Immunotherapy with anti-programmed death 1 (PD-1) or anti-programmed death ligand 1 (PD-L1) monoclonal antibodies has proven to be very effective in the rare group of patients with mCRC harboring deficiencies in mismatch repair, but has unfortunately largely fallen short for the vast majority of patients with mismatch repair proficient cancers. Martinelli et al. looked at adding avelumab, and anti-PD-L1 antibody, to cetuximab in an effort to rechallenge patients with RAS wild-type disease to anti-EGFR therapy in the single-arm phase 2 CAVE trial. 77 patients were treated and the median overall survival was an impressive 11.6 months, increasing to 17.3 months in patients with circulating tumor DNA (ctDNA) that was wild-type for both RAS and BRAF. This study suggests that rechallenging with anti-EGFR therapy can be effective in a ctDNA-selected population and that immunotherapy may have a role to play in this setting, although these findings would need to be confirmed in a larger, randomized phase 3 trial.

Finally, the COVID-19 pandemic has upended medical care, including oncology care. Thierry and co-workers evaluated the effect on the pandemic lockdown in France by looking at the baseline levels of ctDNA in 80 patients with newly diagnosed mCRC presenting pre-lockdown vs. post-lockdown. They showed that median ctDNA was much higher post-lockdown (119.2 vs 17.3 ng/mL), implying that delays in diagnosis related to the pandemic led to more advanced mCRC, at least in terms of ctDNA level. This study adds to the growing literature demonstrating that COVID-19 will continue to have effects on cancer care and cancer-related mortality for years to come.

Author and Disclosure Information

Benjamin A. Weinberg, MD

Disclosures: Speaker/advisor for Bayer, Taiho, Sirtex, HalioDx, Lilly, AstraZeneca/Daiichi Sankyo, research support from Ipsen, G1 Therapeutics.

Publications
Topics
Sections
Author and Disclosure Information

Benjamin A. Weinberg, MD

Disclosures: Speaker/advisor for Bayer, Taiho, Sirtex, HalioDx, Lilly, AstraZeneca/Daiichi Sankyo, research support from Ipsen, G1 Therapeutics.

Author and Disclosure Information

Benjamin A. Weinberg, MD

Disclosures: Speaker/advisor for Bayer, Taiho, Sirtex, HalioDx, Lilly, AstraZeneca/Daiichi Sankyo, research support from Ipsen, G1 Therapeutics.

Dr. Weinberg scans the journals, so you don’t have to!
Dr. Weinberg scans the journals, so you don’t have to!

Benjamin A. Weinberg, MD, FACP
The choice of therapy in patients with refractory metastatic colorectal cancer (mCRC) can be perplexing. Following first- and second-line chemotherapy regimens including fluoropyrimidines, oxaliplatin, irinotecan, and biologics such as bevacizumab (an anti-vascular endothelial growth factor [anti-VEGF] antibody) or cetuximab (an anti-epidermal growth factor receptor [anti-EGFR] antibody), standard later-line options include regorafenib, trifluridine/tipiracil, or rechallenging with chemotherapy and/or an anti-EGFR antibody.

Emerging data have helped to elucidate the real world experience of regorafenib vs trifluridine/tipiracil in the third-line setting. In a single-institution retrospective study, Patel and colleagues evaluated 126 patients who received trifluridine/tipiracil and 95 patients who received regorafenib. There were higher response and disease control rates seen with trifluridine/tipiracil, although median overall survival was similar (7.5 vs 7.1 months). These findings are limited by the single-institution, retrospective and non-randomized nature of the study. Also, the better-tolerated ReDOS dose-escalation schema for regorafenib was not utilized in this study, and it would be interesting to see how this novel dosing strategy compares to trifluridine/tipiracil in the real world setting.

Immunotherapy with anti-programmed death 1 (PD-1) or anti-programmed death ligand 1 (PD-L1) monoclonal antibodies has proven to be very effective in the rare group of patients with mCRC harboring deficiencies in mismatch repair, but has unfortunately largely fallen short for the vast majority of patients with mismatch repair proficient cancers. Martinelli et al. looked at adding avelumab, and anti-PD-L1 antibody, to cetuximab in an effort to rechallenge patients with RAS wild-type disease to anti-EGFR therapy in the single-arm phase 2 CAVE trial. 77 patients were treated and the median overall survival was an impressive 11.6 months, increasing to 17.3 months in patients with circulating tumor DNA (ctDNA) that was wild-type for both RAS and BRAF. This study suggests that rechallenging with anti-EGFR therapy can be effective in a ctDNA-selected population and that immunotherapy may have a role to play in this setting, although these findings would need to be confirmed in a larger, randomized phase 3 trial.

Finally, the COVID-19 pandemic has upended medical care, including oncology care. Thierry and co-workers evaluated the effect on the pandemic lockdown in France by looking at the baseline levels of ctDNA in 80 patients with newly diagnosed mCRC presenting pre-lockdown vs. post-lockdown. They showed that median ctDNA was much higher post-lockdown (119.2 vs 17.3 ng/mL), implying that delays in diagnosis related to the pandemic led to more advanced mCRC, at least in terms of ctDNA level. This study adds to the growing literature demonstrating that COVID-19 will continue to have effects on cancer care and cancer-related mortality for years to come.

Benjamin A. Weinberg, MD, FACP
The choice of therapy in patients with refractory metastatic colorectal cancer (mCRC) can be perplexing. Following first- and second-line chemotherapy regimens including fluoropyrimidines, oxaliplatin, irinotecan, and biologics such as bevacizumab (an anti-vascular endothelial growth factor [anti-VEGF] antibody) or cetuximab (an anti-epidermal growth factor receptor [anti-EGFR] antibody), standard later-line options include regorafenib, trifluridine/tipiracil, or rechallenging with chemotherapy and/or an anti-EGFR antibody.

Emerging data have helped to elucidate the real world experience of regorafenib vs trifluridine/tipiracil in the third-line setting. In a single-institution retrospective study, Patel and colleagues evaluated 126 patients who received trifluridine/tipiracil and 95 patients who received regorafenib. There were higher response and disease control rates seen with trifluridine/tipiracil, although median overall survival was similar (7.5 vs 7.1 months). These findings are limited by the single-institution, retrospective and non-randomized nature of the study. Also, the better-tolerated ReDOS dose-escalation schema for regorafenib was not utilized in this study, and it would be interesting to see how this novel dosing strategy compares to trifluridine/tipiracil in the real world setting.

Immunotherapy with anti-programmed death 1 (PD-1) or anti-programmed death ligand 1 (PD-L1) monoclonal antibodies has proven to be very effective in the rare group of patients with mCRC harboring deficiencies in mismatch repair, but has unfortunately largely fallen short for the vast majority of patients with mismatch repair proficient cancers. Martinelli et al. looked at adding avelumab, and anti-PD-L1 antibody, to cetuximab in an effort to rechallenge patients with RAS wild-type disease to anti-EGFR therapy in the single-arm phase 2 CAVE trial. 77 patients were treated and the median overall survival was an impressive 11.6 months, increasing to 17.3 months in patients with circulating tumor DNA (ctDNA) that was wild-type for both RAS and BRAF. This study suggests that rechallenging with anti-EGFR therapy can be effective in a ctDNA-selected population and that immunotherapy may have a role to play in this setting, although these findings would need to be confirmed in a larger, randomized phase 3 trial.

Finally, the COVID-19 pandemic has upended medical care, including oncology care. Thierry and co-workers evaluated the effect on the pandemic lockdown in France by looking at the baseline levels of ctDNA in 80 patients with newly diagnosed mCRC presenting pre-lockdown vs. post-lockdown. They showed that median ctDNA was much higher post-lockdown (119.2 vs 17.3 ng/mL), implying that delays in diagnosis related to the pandemic led to more advanced mCRC, at least in terms of ctDNA level. This study adds to the growing literature demonstrating that COVID-19 will continue to have effects on cancer care and cancer-related mortality for years to come.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: CRC October 2021
Gate On Date
Fri, 08/27/2021 - 19:30
Un-Gate On Date
Fri, 08/27/2021 - 19:30
Use ProPublica
CFC Schedule Remove Status
Fri, 08/27/2021 - 19:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: Uterine Fibroids October 2021

Article Type
Changed
Fri, 05/13/2022 - 16:34
Dr. Christianson scans the journals, so you don’t have to!

Mindy S. Christianson, MD
The question of the growth of uterine fibroids during menopause was investigated in a recent retrospective longitudinal study published in Menopause (Shen et al). The study team evaluated the medical records of postmenopausal women over a 5-year period. Women received at least two transvaginal ultrasound examinations within a 6-month period. All fibroids were confirmed surgically. Fibroid volume was calculated using the ellipsoid volume formula and the growth rate was calculated. Of 102 postmenopausal women evaluated, the median growth rate was 12.9% every 6 months. Of note, obesity was significantly associated with growth rate (P < 0.05). The estimated growth rate for obese women was 26.6% higher than normal weight women and the growth rate for overweight women was 15.9% higher. Smaller fibroids (< 3 cm diameter) had a higher growth rate than larger uterine fibroids (> 5 cm).

A second study also evaluated the growth uterine fibroids during a different time during a woman’s lifespan - pregnancy. While it’s commonly thought that fibroids grow during pregnancy, a prospective cross-sectional study in the International Journal of Gynecology & Obstetrics (Tian et al) evaluated 394 women with uterine fibroids and found that growth depended on gestational age. In this group, ultrasound examinations were conducted to measure the size of uterine fibroids during weeks 6–7, 11–14, 22–24 and 28–34 of pregnancy and before delivery. The study team found that uterine fibroid size commonly increased before 22–24 weeks of pregnancy with the fastest growth occurring before 11–14 weeks gestation. Later in pregnancy, from 22–24 weeks to the date of delivery, uterine fibroid size remained unchanged.

Lin et al published a large-scale nationwide cohort study in PLoS One that evaluated whether women with uterine fibroids were at increased risk of developing endometriosis. Overall, 31,239 women with uterine fibroids were matched to 124, 956 control participants and followed for 14 years. Compared to controls, patients with uterine fibroids were at a higher risk of developing endometriosis (adjusted hazard ratio [aHR] 6.44, P < 0.05). Other conditions associated with a higher risk of endometriosis included history of tubo-ovarian infection (aHR 2.86, P = 0.01), endometritis (aHR 1.14, P < 0.001), infertility (aHR 1.26, P < 0.001) and allergic diseases (aHR, 1.11, P < .001). Similarly, having both uterine fibroids and infertility significantly increased the risk of endometriosis (aHR 6.95; P < 0.001).

Author and Disclosure Information

Mindy S. Christianson, MD Medical Director, Johns Hopkins Fertility Center
Associate Professor, Division of Reproductive Endocrinology and Infertility
Johns Hopkins University School of Medicine

Dr. Christianson has no disclosures. 

Publications
Topics
Sections
Author and Disclosure Information

Mindy S. Christianson, MD Medical Director, Johns Hopkins Fertility Center
Associate Professor, Division of Reproductive Endocrinology and Infertility
Johns Hopkins University School of Medicine

Dr. Christianson has no disclosures. 

Author and Disclosure Information

Mindy S. Christianson, MD Medical Director, Johns Hopkins Fertility Center
Associate Professor, Division of Reproductive Endocrinology and Infertility
Johns Hopkins University School of Medicine

Dr. Christianson has no disclosures. 

Dr. Christianson scans the journals, so you don’t have to!
Dr. Christianson scans the journals, so you don’t have to!

Mindy S. Christianson, MD
The question of the growth of uterine fibroids during menopause was investigated in a recent retrospective longitudinal study published in Menopause (Shen et al). The study team evaluated the medical records of postmenopausal women over a 5-year period. Women received at least two transvaginal ultrasound examinations within a 6-month period. All fibroids were confirmed surgically. Fibroid volume was calculated using the ellipsoid volume formula and the growth rate was calculated. Of 102 postmenopausal women evaluated, the median growth rate was 12.9% every 6 months. Of note, obesity was significantly associated with growth rate (P < 0.05). The estimated growth rate for obese women was 26.6% higher than normal weight women and the growth rate for overweight women was 15.9% higher. Smaller fibroids (< 3 cm diameter) had a higher growth rate than larger uterine fibroids (> 5 cm).

A second study also evaluated the growth uterine fibroids during a different time during a woman’s lifespan - pregnancy. While it’s commonly thought that fibroids grow during pregnancy, a prospective cross-sectional study in the International Journal of Gynecology & Obstetrics (Tian et al) evaluated 394 women with uterine fibroids and found that growth depended on gestational age. In this group, ultrasound examinations were conducted to measure the size of uterine fibroids during weeks 6–7, 11–14, 22–24 and 28–34 of pregnancy and before delivery. The study team found that uterine fibroid size commonly increased before 22–24 weeks of pregnancy with the fastest growth occurring before 11–14 weeks gestation. Later in pregnancy, from 22–24 weeks to the date of delivery, uterine fibroid size remained unchanged.

Lin et al published a large-scale nationwide cohort study in PLoS One that evaluated whether women with uterine fibroids were at increased risk of developing endometriosis. Overall, 31,239 women with uterine fibroids were matched to 124, 956 control participants and followed for 14 years. Compared to controls, patients with uterine fibroids were at a higher risk of developing endometriosis (adjusted hazard ratio [aHR] 6.44, P < 0.05). Other conditions associated with a higher risk of endometriosis included history of tubo-ovarian infection (aHR 2.86, P = 0.01), endometritis (aHR 1.14, P < 0.001), infertility (aHR 1.26, P < 0.001) and allergic diseases (aHR, 1.11, P < .001). Similarly, having both uterine fibroids and infertility significantly increased the risk of endometriosis (aHR 6.95; P < 0.001).

Mindy S. Christianson, MD
The question of the growth of uterine fibroids during menopause was investigated in a recent retrospective longitudinal study published in Menopause (Shen et al). The study team evaluated the medical records of postmenopausal women over a 5-year period. Women received at least two transvaginal ultrasound examinations within a 6-month period. All fibroids were confirmed surgically. Fibroid volume was calculated using the ellipsoid volume formula and the growth rate was calculated. Of 102 postmenopausal women evaluated, the median growth rate was 12.9% every 6 months. Of note, obesity was significantly associated with growth rate (P < 0.05). The estimated growth rate for obese women was 26.6% higher than normal weight women and the growth rate for overweight women was 15.9% higher. Smaller fibroids (< 3 cm diameter) had a higher growth rate than larger uterine fibroids (> 5 cm).

A second study also evaluated the growth uterine fibroids during a different time during a woman’s lifespan - pregnancy. While it’s commonly thought that fibroids grow during pregnancy, a prospective cross-sectional study in the International Journal of Gynecology & Obstetrics (Tian et al) evaluated 394 women with uterine fibroids and found that growth depended on gestational age. In this group, ultrasound examinations were conducted to measure the size of uterine fibroids during weeks 6–7, 11–14, 22–24 and 28–34 of pregnancy and before delivery. The study team found that uterine fibroid size commonly increased before 22–24 weeks of pregnancy with the fastest growth occurring before 11–14 weeks gestation. Later in pregnancy, from 22–24 weeks to the date of delivery, uterine fibroid size remained unchanged.

Lin et al published a large-scale nationwide cohort study in PLoS One that evaluated whether women with uterine fibroids were at increased risk of developing endometriosis. Overall, 31,239 women with uterine fibroids were matched to 124, 956 control participants and followed for 14 years. Compared to controls, patients with uterine fibroids were at a higher risk of developing endometriosis (adjusted hazard ratio [aHR] 6.44, P < 0.05). Other conditions associated with a higher risk of endometriosis included history of tubo-ovarian infection (aHR 2.86, P = 0.01), endometritis (aHR 1.14, P < 0.001), infertility (aHR 1.26, P < 0.001) and allergic diseases (aHR, 1.11, P < .001). Similarly, having both uterine fibroids and infertility significantly increased the risk of endometriosis (aHR 6.95; P < 0.001).

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Uterine Fibroids October 2021
Gate On Date
Thu, 07/29/2021 - 18:45
Un-Gate On Date
Thu, 07/29/2021 - 18:45
Use ProPublica
CFC Schedule Remove Status
Thu, 07/29/2021 - 18:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: CML October 2021

Article Type
Changed
Fri, 05/06/2022 - 16:50
Dr. Pinilla-Ibarz scans the journals, so you don’t have to!

Javier Pinilla-Ibarz MD, PhD

Ponatinib is most likely the most powerful tyrosine kinase inhibitor (TKI) approved for the relapsed or refractory chronic myeloid leukemia (CML) after two lines of therapy. In the original PACE study, ponatinib showed deep and durable responses, but arterial occlusive events (AOE) emerged as notable adverse events. Post hoc analyses indicated that AOEs were dose dependent and recommendations to lower the dose from 45 mg to 30 mg after achieving complete cytogenic response (CCR) and to 15 mg after achieving major molecular response (MMR) were used in an attempt to minimize the cardiovascular toxicities. The question remained regarding the optimal dose of ponatinib in terms of efficacy and toxicity. Cortes et al (Blood 2021 Aug 18.) reported the results of the OPTIC trial where patients with chronic phase CML (CP-CML), resistant/intolerant to at least 2 prior BCR-ABL1 TKIs or with a BCR-ABL1T315I mutation, were randomized 1:1:1 to receive 3 different doses of ponatinib daily (45, 30, or 15 mg). Patients who received 45 mg or 30 mg daily reduced their dose to 15 mg upon achievement of response (BCR-ABL1IS transcript levels ≤1%). The primary endpoint (BCR-ABL1IS transcript levels ≤1%) was achieved at 12 months in 44.1%, 29.0%, and 23.1% in the 3 cohorts, respectively. Independently confirmed grade 3/4 treatment-emergent AOEs occurred in 5, 5, and 3 patients in the 3 cohorts, respectively. In conclusion, the optimal benefit:risk outcomes occurred with the 45 mg starting dose reduced to 15 mg upon achievement of response.

 

New drugs with alternative mechanisms of action are always welcome for the treatment of resistant or intolerant CP-CML. Asciminib is a first-in-class STAMP (Specifically Targeting the ABL Myristoyl Pocket) inhibitor with the potential to overcome resistance or intolerance to approved TKIs. Rea et al (Blood 2021 Aug 18) reported the results of the ASCEMBL trial that compare 40 mg twice daily asciminib vs. bosutinib in patients with CP-CML refractory to >2 previous lines of therapy. The rates of MMR at week 24 in patients receiving asciminib and bosutinib were 25.5% and 13.2%, respectively (difference 12.2%; P = .029). The bosutinib arm, compared to the asciminib arm, had a more frequent occurrence of grade 3 or higher adverse events (AE; 60.5% vs. 50.6%) and AEs leading to treatment discontinuation (21.1% vs. 5.8%). This will be another important alternative for CP-CML patients failing 2 lines of therapy; however, it is still unclear if the drug may be superior to ponatinib in the same setting.

 

In the new era of COVID-19, patients with hematologic malignancies are at an increased risk of SARS-CoV-2 disease (COVID-19) and an adverse outcome. However, is unclear if this can be seen across the board in all type of hematologic malignancies, as a low mortality rate has been described in patients with CP-CML, suggesting that TKIs may have a protective role against severe COVID-19. Bonifacio et al (Cancer Med 2021 Aug 31) conducted a cross-sectional study of 564 consecutive patients with CML who were tested for anti-SARS-CoV-2 IgG/IgM antibodies at their first outpatient visit between May and early November 2020 in 5 hematologic centers representative of 3 Italian regions. Interestingly, the serological prevalence of SARS-CoV-2 infection in patients with CML after the first pandemic wave was similar to that in the general population. The data confirm mild SARS-CoV-2 infection in patients with CML and suggest that patients with CML succeed to mount an antibody response after exposure to SARS-CoV-2 similar to the general population.

Author and Disclosure Information

Javier Pinilla-Ibarz MD, PhD, Senior Member, Lymphoma Section Head and Director of Immunotherapy, Malignant Hematology Department, H.Lee Moffitt Cancer Center & Research Institute

Javier Pinilla has disclosed the following relevant financial relationships:

Serve(d) as a speaker or a member of a speakers bureau for: Abbvie; Janssen; Takeda; AstraZeneca
Received research grant from: TG therapeutics; MEI; Sunesis
Publications
Topics
Sections
Author and Disclosure Information

Javier Pinilla-Ibarz MD, PhD, Senior Member, Lymphoma Section Head and Director of Immunotherapy, Malignant Hematology Department, H.Lee Moffitt Cancer Center & Research Institute

Javier Pinilla has disclosed the following relevant financial relationships:

Serve(d) as a speaker or a member of a speakers bureau for: Abbvie; Janssen; Takeda; AstraZeneca
Received research grant from: TG therapeutics; MEI; Sunesis
Author and Disclosure Information

Javier Pinilla-Ibarz MD, PhD, Senior Member, Lymphoma Section Head and Director of Immunotherapy, Malignant Hematology Department, H.Lee Moffitt Cancer Center & Research Institute

Javier Pinilla has disclosed the following relevant financial relationships:

Serve(d) as a speaker or a member of a speakers bureau for: Abbvie; Janssen; Takeda; AstraZeneca
Received research grant from: TG therapeutics; MEI; Sunesis
Dr. Pinilla-Ibarz scans the journals, so you don’t have to!
Dr. Pinilla-Ibarz scans the journals, so you don’t have to!

Javier Pinilla-Ibarz MD, PhD

Ponatinib is most likely the most powerful tyrosine kinase inhibitor (TKI) approved for the relapsed or refractory chronic myeloid leukemia (CML) after two lines of therapy. In the original PACE study, ponatinib showed deep and durable responses, but arterial occlusive events (AOE) emerged as notable adverse events. Post hoc analyses indicated that AOEs were dose dependent and recommendations to lower the dose from 45 mg to 30 mg after achieving complete cytogenic response (CCR) and to 15 mg after achieving major molecular response (MMR) were used in an attempt to minimize the cardiovascular toxicities. The question remained regarding the optimal dose of ponatinib in terms of efficacy and toxicity. Cortes et al (Blood 2021 Aug 18.) reported the results of the OPTIC trial where patients with chronic phase CML (CP-CML), resistant/intolerant to at least 2 prior BCR-ABL1 TKIs or with a BCR-ABL1T315I mutation, were randomized 1:1:1 to receive 3 different doses of ponatinib daily (45, 30, or 15 mg). Patients who received 45 mg or 30 mg daily reduced their dose to 15 mg upon achievement of response (BCR-ABL1IS transcript levels ≤1%). The primary endpoint (BCR-ABL1IS transcript levels ≤1%) was achieved at 12 months in 44.1%, 29.0%, and 23.1% in the 3 cohorts, respectively. Independently confirmed grade 3/4 treatment-emergent AOEs occurred in 5, 5, and 3 patients in the 3 cohorts, respectively. In conclusion, the optimal benefit:risk outcomes occurred with the 45 mg starting dose reduced to 15 mg upon achievement of response.

 

New drugs with alternative mechanisms of action are always welcome for the treatment of resistant or intolerant CP-CML. Asciminib is a first-in-class STAMP (Specifically Targeting the ABL Myristoyl Pocket) inhibitor with the potential to overcome resistance or intolerance to approved TKIs. Rea et al (Blood 2021 Aug 18) reported the results of the ASCEMBL trial that compare 40 mg twice daily asciminib vs. bosutinib in patients with CP-CML refractory to >2 previous lines of therapy. The rates of MMR at week 24 in patients receiving asciminib and bosutinib were 25.5% and 13.2%, respectively (difference 12.2%; P = .029). The bosutinib arm, compared to the asciminib arm, had a more frequent occurrence of grade 3 or higher adverse events (AE; 60.5% vs. 50.6%) and AEs leading to treatment discontinuation (21.1% vs. 5.8%). This will be another important alternative for CP-CML patients failing 2 lines of therapy; however, it is still unclear if the drug may be superior to ponatinib in the same setting.

 

In the new era of COVID-19, patients with hematologic malignancies are at an increased risk of SARS-CoV-2 disease (COVID-19) and an adverse outcome. However, is unclear if this can be seen across the board in all type of hematologic malignancies, as a low mortality rate has been described in patients with CP-CML, suggesting that TKIs may have a protective role against severe COVID-19. Bonifacio et al (Cancer Med 2021 Aug 31) conducted a cross-sectional study of 564 consecutive patients with CML who were tested for anti-SARS-CoV-2 IgG/IgM antibodies at their first outpatient visit between May and early November 2020 in 5 hematologic centers representative of 3 Italian regions. Interestingly, the serological prevalence of SARS-CoV-2 infection in patients with CML after the first pandemic wave was similar to that in the general population. The data confirm mild SARS-CoV-2 infection in patients with CML and suggest that patients with CML succeed to mount an antibody response after exposure to SARS-CoV-2 similar to the general population.

Javier Pinilla-Ibarz MD, PhD

Ponatinib is most likely the most powerful tyrosine kinase inhibitor (TKI) approved for the relapsed or refractory chronic myeloid leukemia (CML) after two lines of therapy. In the original PACE study, ponatinib showed deep and durable responses, but arterial occlusive events (AOE) emerged as notable adverse events. Post hoc analyses indicated that AOEs were dose dependent and recommendations to lower the dose from 45 mg to 30 mg after achieving complete cytogenic response (CCR) and to 15 mg after achieving major molecular response (MMR) were used in an attempt to minimize the cardiovascular toxicities. The question remained regarding the optimal dose of ponatinib in terms of efficacy and toxicity. Cortes et al (Blood 2021 Aug 18.) reported the results of the OPTIC trial where patients with chronic phase CML (CP-CML), resistant/intolerant to at least 2 prior BCR-ABL1 TKIs or with a BCR-ABL1T315I mutation, were randomized 1:1:1 to receive 3 different doses of ponatinib daily (45, 30, or 15 mg). Patients who received 45 mg or 30 mg daily reduced their dose to 15 mg upon achievement of response (BCR-ABL1IS transcript levels ≤1%). The primary endpoint (BCR-ABL1IS transcript levels ≤1%) was achieved at 12 months in 44.1%, 29.0%, and 23.1% in the 3 cohorts, respectively. Independently confirmed grade 3/4 treatment-emergent AOEs occurred in 5, 5, and 3 patients in the 3 cohorts, respectively. In conclusion, the optimal benefit:risk outcomes occurred with the 45 mg starting dose reduced to 15 mg upon achievement of response.

 

New drugs with alternative mechanisms of action are always welcome for the treatment of resistant or intolerant CP-CML. Asciminib is a first-in-class STAMP (Specifically Targeting the ABL Myristoyl Pocket) inhibitor with the potential to overcome resistance or intolerance to approved TKIs. Rea et al (Blood 2021 Aug 18) reported the results of the ASCEMBL trial that compare 40 mg twice daily asciminib vs. bosutinib in patients with CP-CML refractory to >2 previous lines of therapy. The rates of MMR at week 24 in patients receiving asciminib and bosutinib were 25.5% and 13.2%, respectively (difference 12.2%; P = .029). The bosutinib arm, compared to the asciminib arm, had a more frequent occurrence of grade 3 or higher adverse events (AE; 60.5% vs. 50.6%) and AEs leading to treatment discontinuation (21.1% vs. 5.8%). This will be another important alternative for CP-CML patients failing 2 lines of therapy; however, it is still unclear if the drug may be superior to ponatinib in the same setting.

 

In the new era of COVID-19, patients with hematologic malignancies are at an increased risk of SARS-CoV-2 disease (COVID-19) and an adverse outcome. However, is unclear if this can be seen across the board in all type of hematologic malignancies, as a low mortality rate has been described in patients with CP-CML, suggesting that TKIs may have a protective role against severe COVID-19. Bonifacio et al (Cancer Med 2021 Aug 31) conducted a cross-sectional study of 564 consecutive patients with CML who were tested for anti-SARS-CoV-2 IgG/IgM antibodies at their first outpatient visit between May and early November 2020 in 5 hematologic centers representative of 3 Italian regions. Interestingly, the serological prevalence of SARS-CoV-2 infection in patients with CML after the first pandemic wave was similar to that in the general population. The data confirm mild SARS-CoV-2 infection in patients with CML and suggest that patients with CML succeed to mount an antibody response after exposure to SARS-CoV-2 similar to the general population.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: CML October 2021
Gate On Date
Tue, 04/06/2021 - 09:00
Un-Gate On Date
Tue, 04/06/2021 - 09:00
Use ProPublica
CFC Schedule Remove Status
Tue, 04/06/2021 - 09:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: Atopic Dermatitis October 2021

Article Type
Changed
Thu, 05/12/2022 - 11:42
Dr. Silverberg scans the journals, so you don’t have to!

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC
Atopic dermatitis management is getting JAK’d up

Topical and oral Janus Kinase (JAK)-inhibitors are important new additions to the therapeutic armamentarium of atopic dermatitis (AD). I recently addressed some important treatment considerations regarding the JAK-inhibitors. In just two short months, there have already been a number of important new publications on JAK-inhibitors in AD that provide crucial data to guide treatment decisions.

Topical ruxolitinib 1.5% cream (a JAK1/2 inhibitor) was just approved by United States Food and Drug Administration for the treatment of mild-moderate AD. Clinicians always want to know about the comparative effectiveness of new agents compared to already approved agents. A previous phase 2B randomized controlled trial (RCT) compared multiple doses of ruxolitinib cream with a vehicle control and triamcinolone 0.1% cream active comparator1. Topical ruxolitinib 1.5% cream was significantly more effective than vehicle and numerically more effective than triamcinolone 0.1% cream.

Zhang et al. recently conducted a network meta-analysis of 10 RCT for topical JAK and phosphodiesterase E4 (PDE4)-inhibitors, mostly with mild-to-moderate AD. All included JAK inhibitors showed higher Investigators Global Assessment (IGA) response vs. vehicle, with ruxolitinib 1.5% once daily showing similar efficacy as tofacitinib 2% and delgocitinib 3% twice daily. Whereas, topical tacrolimus 0.1% and hydrocortisone butyrate 0.1% twice a day were not more effective than vehicle at achieving IGA response. These results suggest that topical ruxolitinib and other JAK-inhibitors are more effective at clearing AD lesions than currently used topical therapies.

There has been a recent flurry of publications regarding the efficacy and safety of abrocitinib (an oral, once daily, JAK1 inhibitor) in moderate-severe atopic dermatitis.

  • Eichenfield et al. published the results of the JADE TEEN study 2, a phase 3 RCT of abrocitinib in adolescents. Abrocitinib 200 mg and 100 mg resulted in significant improvements of IGA, Eczema Area and Severity Index, and itch scores, etc. over a 12-week treatment period compared to placebo. These results support the efficacy of abrocitinib in adolescents with moderate-severe AD.
  • Simpson et al. published the results from an integrated safety analysis of pooled data from 5 short-term and 1 long-term extension study of abrocitinib therapy 3. Abrocitinib 200 mg and 100 mg doses were well-tolerated during 12-week placebo controlled trials, with nausea, headache, and acne being the most common adverse-events. The incidence of different adverse-events did not consistently increase over time. However, there were some rare events reported for venous thromboembolism and deaths. These results indicate an overall good safety profile for abrocitinib, but proper patient and dose selection should be carefully considered.
  • Additionally, strategies should be employed to potentially minimize risk of adverse-events. One such approach is flexible dosing in order to maintain long-term disease control using the lowest amount of medicine needed. Blauvelt et al. published findings from the JADE REGIMEN study 4. Patients who responded to 12 weeks of abrocitinib 200 mg open-label monotherapy were randomly assigned to abrocitinib 200 mg, abrocitinib 100 mg, or placebo maintenance therapy for 40-weeks. Flares occurred least commonly in patients maintained on abrocitinib 200 mg (18.9%), followed by abrocitinib 100 mg (42.6%), and most commonly for placebo (80.9%). These results indicate that a large subset of patients who achieve clinical response with abrocitinib 200 mg could be maintained on a lower dose of 100 mg and in some cases may even be able to have a drug holiday without flaring. While similar studies were not performed for other oral JAK-inhibitors, it may be that lower maintenance dosing may also be feasible and effective for other oral JAK-inhibitors. Future research is needed to identify patient subsets who will most likely maintain clinical response with lower maintenance dosing of oral JAK-inhibitors.
  1. Kim BS, Howell MD, Sun K, et al. Treatment of atopic dermatitis with ruxolitinib cream (JAK1/JAK2 inhibitor) or triamcinolone cream. The Journal of allergy and clinical immunology. 2020;145(2):572-582.
  2. Eichenfield LF, Flohr C, Sidbury R, et al. Efficacy and Safety of Abrocitinib in Combination With Topical Therapy in Adolescents With Moderate-to-Severe Atopic Dermatitis: The JADE TEEN Randomized Clinical Trial. JAMA dermatology. 2021.
  3. Simpson EL, Silverberg JI, Nosbaum A, et al. Integrated Safety Analysis of Abrocitinib for the Treatment of Moderate-to-Severe Atopic Dermatitis From the Phase II and Phase III Clinical Trial Program. American journal of clinical dermatology. 2021;22(5):693-707.
  4. Blauvelt A, Silverberg JI, Lynde CW, et al. Abrocitinib induction, randomized withdrawal, and retreatment in patients with moderate-to-severe atopic dermatitis: Results from the JAK1 Atopic Dermatitis Efficacy and Safety (JADE) REGIMEN phase 3 trial. Journal of the American Academy of Dermatology.
Author and Disclosure Information

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC

Publications
Topics
Sections
Author and Disclosure Information

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC

Author and Disclosure Information

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC

Dr. Silverberg scans the journals, so you don’t have to!
Dr. Silverberg scans the journals, so you don’t have to!

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC
Atopic dermatitis management is getting JAK’d up

Topical and oral Janus Kinase (JAK)-inhibitors are important new additions to the therapeutic armamentarium of atopic dermatitis (AD). I recently addressed some important treatment considerations regarding the JAK-inhibitors. In just two short months, there have already been a number of important new publications on JAK-inhibitors in AD that provide crucial data to guide treatment decisions.

Topical ruxolitinib 1.5% cream (a JAK1/2 inhibitor) was just approved by United States Food and Drug Administration for the treatment of mild-moderate AD. Clinicians always want to know about the comparative effectiveness of new agents compared to already approved agents. A previous phase 2B randomized controlled trial (RCT) compared multiple doses of ruxolitinib cream with a vehicle control and triamcinolone 0.1% cream active comparator1. Topical ruxolitinib 1.5% cream was significantly more effective than vehicle and numerically more effective than triamcinolone 0.1% cream.

Zhang et al. recently conducted a network meta-analysis of 10 RCT for topical JAK and phosphodiesterase E4 (PDE4)-inhibitors, mostly with mild-to-moderate AD. All included JAK inhibitors showed higher Investigators Global Assessment (IGA) response vs. vehicle, with ruxolitinib 1.5% once daily showing similar efficacy as tofacitinib 2% and delgocitinib 3% twice daily. Whereas, topical tacrolimus 0.1% and hydrocortisone butyrate 0.1% twice a day were not more effective than vehicle at achieving IGA response. These results suggest that topical ruxolitinib and other JAK-inhibitors are more effective at clearing AD lesions than currently used topical therapies.

There has been a recent flurry of publications regarding the efficacy and safety of abrocitinib (an oral, once daily, JAK1 inhibitor) in moderate-severe atopic dermatitis.

  • Eichenfield et al. published the results of the JADE TEEN study 2, a phase 3 RCT of abrocitinib in adolescents. Abrocitinib 200 mg and 100 mg resulted in significant improvements of IGA, Eczema Area and Severity Index, and itch scores, etc. over a 12-week treatment period compared to placebo. These results support the efficacy of abrocitinib in adolescents with moderate-severe AD.
  • Simpson et al. published the results from an integrated safety analysis of pooled data from 5 short-term and 1 long-term extension study of abrocitinib therapy 3. Abrocitinib 200 mg and 100 mg doses were well-tolerated during 12-week placebo controlled trials, with nausea, headache, and acne being the most common adverse-events. The incidence of different adverse-events did not consistently increase over time. However, there were some rare events reported for venous thromboembolism and deaths. These results indicate an overall good safety profile for abrocitinib, but proper patient and dose selection should be carefully considered.
  • Additionally, strategies should be employed to potentially minimize risk of adverse-events. One such approach is flexible dosing in order to maintain long-term disease control using the lowest amount of medicine needed. Blauvelt et al. published findings from the JADE REGIMEN study 4. Patients who responded to 12 weeks of abrocitinib 200 mg open-label monotherapy were randomly assigned to abrocitinib 200 mg, abrocitinib 100 mg, or placebo maintenance therapy for 40-weeks. Flares occurred least commonly in patients maintained on abrocitinib 200 mg (18.9%), followed by abrocitinib 100 mg (42.6%), and most commonly for placebo (80.9%). These results indicate that a large subset of patients who achieve clinical response with abrocitinib 200 mg could be maintained on a lower dose of 100 mg and in some cases may even be able to have a drug holiday without flaring. While similar studies were not performed for other oral JAK-inhibitors, it may be that lower maintenance dosing may also be feasible and effective for other oral JAK-inhibitors. Future research is needed to identify patient subsets who will most likely maintain clinical response with lower maintenance dosing of oral JAK-inhibitors.
  1. Kim BS, Howell MD, Sun K, et al. Treatment of atopic dermatitis with ruxolitinib cream (JAK1/JAK2 inhibitor) or triamcinolone cream. The Journal of allergy and clinical immunology. 2020;145(2):572-582.
  2. Eichenfield LF, Flohr C, Sidbury R, et al. Efficacy and Safety of Abrocitinib in Combination With Topical Therapy in Adolescents With Moderate-to-Severe Atopic Dermatitis: The JADE TEEN Randomized Clinical Trial. JAMA dermatology. 2021.
  3. Simpson EL, Silverberg JI, Nosbaum A, et al. Integrated Safety Analysis of Abrocitinib for the Treatment of Moderate-to-Severe Atopic Dermatitis From the Phase II and Phase III Clinical Trial Program. American journal of clinical dermatology. 2021;22(5):693-707.
  4. Blauvelt A, Silverberg JI, Lynde CW, et al. Abrocitinib induction, randomized withdrawal, and retreatment in patients with moderate-to-severe atopic dermatitis: Results from the JAK1 Atopic Dermatitis Efficacy and Safety (JADE) REGIMEN phase 3 trial. Journal of the American Academy of Dermatology.

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC
Atopic dermatitis management is getting JAK’d up

Topical and oral Janus Kinase (JAK)-inhibitors are important new additions to the therapeutic armamentarium of atopic dermatitis (AD). I recently addressed some important treatment considerations regarding the JAK-inhibitors. In just two short months, there have already been a number of important new publications on JAK-inhibitors in AD that provide crucial data to guide treatment decisions.

Topical ruxolitinib 1.5% cream (a JAK1/2 inhibitor) was just approved by United States Food and Drug Administration for the treatment of mild-moderate AD. Clinicians always want to know about the comparative effectiveness of new agents compared to already approved agents. A previous phase 2B randomized controlled trial (RCT) compared multiple doses of ruxolitinib cream with a vehicle control and triamcinolone 0.1% cream active comparator1. Topical ruxolitinib 1.5% cream was significantly more effective than vehicle and numerically more effective than triamcinolone 0.1% cream.

Zhang et al. recently conducted a network meta-analysis of 10 RCT for topical JAK and phosphodiesterase E4 (PDE4)-inhibitors, mostly with mild-to-moderate AD. All included JAK inhibitors showed higher Investigators Global Assessment (IGA) response vs. vehicle, with ruxolitinib 1.5% once daily showing similar efficacy as tofacitinib 2% and delgocitinib 3% twice daily. Whereas, topical tacrolimus 0.1% and hydrocortisone butyrate 0.1% twice a day were not more effective than vehicle at achieving IGA response. These results suggest that topical ruxolitinib and other JAK-inhibitors are more effective at clearing AD lesions than currently used topical therapies.

There has been a recent flurry of publications regarding the efficacy and safety of abrocitinib (an oral, once daily, JAK1 inhibitor) in moderate-severe atopic dermatitis.

  • Eichenfield et al. published the results of the JADE TEEN study 2, a phase 3 RCT of abrocitinib in adolescents. Abrocitinib 200 mg and 100 mg resulted in significant improvements of IGA, Eczema Area and Severity Index, and itch scores, etc. over a 12-week treatment period compared to placebo. These results support the efficacy of abrocitinib in adolescents with moderate-severe AD.
  • Simpson et al. published the results from an integrated safety analysis of pooled data from 5 short-term and 1 long-term extension study of abrocitinib therapy 3. Abrocitinib 200 mg and 100 mg doses were well-tolerated during 12-week placebo controlled trials, with nausea, headache, and acne being the most common adverse-events. The incidence of different adverse-events did not consistently increase over time. However, there were some rare events reported for venous thromboembolism and deaths. These results indicate an overall good safety profile for abrocitinib, but proper patient and dose selection should be carefully considered.
  • Additionally, strategies should be employed to potentially minimize risk of adverse-events. One such approach is flexible dosing in order to maintain long-term disease control using the lowest amount of medicine needed. Blauvelt et al. published findings from the JADE REGIMEN study 4. Patients who responded to 12 weeks of abrocitinib 200 mg open-label monotherapy were randomly assigned to abrocitinib 200 mg, abrocitinib 100 mg, or placebo maintenance therapy for 40-weeks. Flares occurred least commonly in patients maintained on abrocitinib 200 mg (18.9%), followed by abrocitinib 100 mg (42.6%), and most commonly for placebo (80.9%). These results indicate that a large subset of patients who achieve clinical response with abrocitinib 200 mg could be maintained on a lower dose of 100 mg and in some cases may even be able to have a drug holiday without flaring. While similar studies were not performed for other oral JAK-inhibitors, it may be that lower maintenance dosing may also be feasible and effective for other oral JAK-inhibitors. Future research is needed to identify patient subsets who will most likely maintain clinical response with lower maintenance dosing of oral JAK-inhibitors.
  1. Kim BS, Howell MD, Sun K, et al. Treatment of atopic dermatitis with ruxolitinib cream (JAK1/JAK2 inhibitor) or triamcinolone cream. The Journal of allergy and clinical immunology. 2020;145(2):572-582.
  2. Eichenfield LF, Flohr C, Sidbury R, et al. Efficacy and Safety of Abrocitinib in Combination With Topical Therapy in Adolescents With Moderate-to-Severe Atopic Dermatitis: The JADE TEEN Randomized Clinical Trial. JAMA dermatology. 2021.
  3. Simpson EL, Silverberg JI, Nosbaum A, et al. Integrated Safety Analysis of Abrocitinib for the Treatment of Moderate-to-Severe Atopic Dermatitis From the Phase II and Phase III Clinical Trial Program. American journal of clinical dermatology. 2021;22(5):693-707.
  4. Blauvelt A, Silverberg JI, Lynde CW, et al. Abrocitinib induction, randomized withdrawal, and retreatment in patients with moderate-to-severe atopic dermatitis: Results from the JAK1 Atopic Dermatitis Efficacy and Safety (JADE) REGIMEN phase 3 trial. Journal of the American Academy of Dermatology.
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Atopic Dermatitis October 2021
Gate On Date
Thu, 07/29/2021 - 18:45
Un-Gate On Date
Thu, 07/29/2021 - 18:45
Use ProPublica
CFC Schedule Remove Status
Thu, 07/29/2021 - 18:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: Psoriasis October 2021

Article Type
Changed
Thu, 05/12/2022 - 11:42
Dr. Ferris scans the journals, so you don’t have to!

Laura Ferris, MD, PhD
While biologics have revolutionized the treatment of psoriasis, oral and topical therapies remain excellent options for many patients. Several recent studies highlight new data on non-biologic therapies.

High-potency topical steroids such as clobetasol are commonly used as first-line treatment for psoriasis. A recent study (Sidgiddi S et al. Dermatol Ther (Heidelb). 2021 Aug 28) compared the efficacy and safety of clobetasol 0.05% vs. 0.025% cream.  Patients were randomized to receive either clobetasol 0.05% cream or one of two different formulations of clobetasol 0.025% cream twice a day for two weeks. PGA success rates (clear or almost clear skin) were higher with the 0.025% formulations (38.9% and 36.8%) than with the 0.05% cream (30.8%). Safety also appeared to be better or comparable with the 0.025% formulation as measured by the proportion of patients with an abnormal ACTH stimulation test (20.7% and 17.2% in the 0.025% group compared with 30.0% in the 0.05% group). Due to the small study size (88 subjects) these differences did not reach statistical significance, although they suggest that high efficacy and perhaps better safety (reduced hypothalamic–pituitary–adrenal axis suppression) can be achieved with lower concentration formulations of clobetasol cream.

The oral phosphodiesterase 4 inhibitor apremilast is FDA-approved to treat psoriasis and psoriatic arthritis and recent studies have shown that it is more effective than placebo in treating patients with mild-moderate psoriasis and scalp psoriasis. A recent prospective cohort study followed 45 adult patients with plaque and nail psoriasis with a fingernail Nail Psoriasis Severity Index (NAPSI) score of 12 or more treated with apremilast 30 mg twice daily for 52 weeks (Muñoz-Santos C et al. J Dermatol. 2021 Aug 12). The primary endpoint, the percentage of patients with a Nail Assessment in Psoriasis and Psoriatic Arthritis-Patient Benefit Index of 2 or more at week 52, was achieved in 52% of patients. A median improvement of 53% in fingernail NAPSI score and a mean reduction in nail pain of 68% were observed at week 52.  These findings show that apremilast can be useful in improving the quality-of-life impairment caused by nail psoriasis.

Traditional systemic therapies such as methotrexate and acitretin are cost-effective options for many psoriasis patients. The association of psoriasis with other comorbid conditions, particularly cardiovascular disease, has raised awareness of the importance of considering how therapies impact not just skin disease but also the risk posed by these psoriasis-associated comorbidities. A large Taiwanese retrospective cohort study compared patients treated with methotrexate (13,777) or acitretin (6,020) and found that in comparison to those treated with acitretin, patients treated with methotrexate were at lower risk of experiencing adverse cardiovascular outcomes, including ischemic heart disease and stroke, (adjusted hazard ratio [aHR], 0.84; 95% confidence interval [CI], 0.76-0.94) and had a lower risk of all-cause mortality (aHR, 0.75; 95% CI, 0.66-0.85).

Many disease and patient factors must be considered when choosing the right therapy for a patient. These studies provide valuable information to incorporate into this process and highlight the utility of topical and oral therapies for psoriasis.

Author and Disclosure Information

Laura K. Ferris, MD, PhD, Professor, Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania

Laura K. Ferris, MD, PhD, has disclosed the following relevant financial relationships:

Received income in an amount equal to or greater than $250 from: Eli Lilly; Janssen; Bristol-Myers Squibb; Arcutis; Dermavant; AbbVie

Publications
Topics
Sections
Author and Disclosure Information

Laura K. Ferris, MD, PhD, Professor, Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania

Laura K. Ferris, MD, PhD, has disclosed the following relevant financial relationships:

Received income in an amount equal to or greater than $250 from: Eli Lilly; Janssen; Bristol-Myers Squibb; Arcutis; Dermavant; AbbVie

Author and Disclosure Information

Laura K. Ferris, MD, PhD, Professor, Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania

Laura K. Ferris, MD, PhD, has disclosed the following relevant financial relationships:

Received income in an amount equal to or greater than $250 from: Eli Lilly; Janssen; Bristol-Myers Squibb; Arcutis; Dermavant; AbbVie

Dr. Ferris scans the journals, so you don’t have to!
Dr. Ferris scans the journals, so you don’t have to!

Laura Ferris, MD, PhD
While biologics have revolutionized the treatment of psoriasis, oral and topical therapies remain excellent options for many patients. Several recent studies highlight new data on non-biologic therapies.

High-potency topical steroids such as clobetasol are commonly used as first-line treatment for psoriasis. A recent study (Sidgiddi S et al. Dermatol Ther (Heidelb). 2021 Aug 28) compared the efficacy and safety of clobetasol 0.05% vs. 0.025% cream.  Patients were randomized to receive either clobetasol 0.05% cream or one of two different formulations of clobetasol 0.025% cream twice a day for two weeks. PGA success rates (clear or almost clear skin) were higher with the 0.025% formulations (38.9% and 36.8%) than with the 0.05% cream (30.8%). Safety also appeared to be better or comparable with the 0.025% formulation as measured by the proportion of patients with an abnormal ACTH stimulation test (20.7% and 17.2% in the 0.025% group compared with 30.0% in the 0.05% group). Due to the small study size (88 subjects) these differences did not reach statistical significance, although they suggest that high efficacy and perhaps better safety (reduced hypothalamic–pituitary–adrenal axis suppression) can be achieved with lower concentration formulations of clobetasol cream.

The oral phosphodiesterase 4 inhibitor apremilast is FDA-approved to treat psoriasis and psoriatic arthritis and recent studies have shown that it is more effective than placebo in treating patients with mild-moderate psoriasis and scalp psoriasis. A recent prospective cohort study followed 45 adult patients with plaque and nail psoriasis with a fingernail Nail Psoriasis Severity Index (NAPSI) score of 12 or more treated with apremilast 30 mg twice daily for 52 weeks (Muñoz-Santos C et al. J Dermatol. 2021 Aug 12). The primary endpoint, the percentage of patients with a Nail Assessment in Psoriasis and Psoriatic Arthritis-Patient Benefit Index of 2 or more at week 52, was achieved in 52% of patients. A median improvement of 53% in fingernail NAPSI score and a mean reduction in nail pain of 68% were observed at week 52.  These findings show that apremilast can be useful in improving the quality-of-life impairment caused by nail psoriasis.

Traditional systemic therapies such as methotrexate and acitretin are cost-effective options for many psoriasis patients. The association of psoriasis with other comorbid conditions, particularly cardiovascular disease, has raised awareness of the importance of considering how therapies impact not just skin disease but also the risk posed by these psoriasis-associated comorbidities. A large Taiwanese retrospective cohort study compared patients treated with methotrexate (13,777) or acitretin (6,020) and found that in comparison to those treated with acitretin, patients treated with methotrexate were at lower risk of experiencing adverse cardiovascular outcomes, including ischemic heart disease and stroke, (adjusted hazard ratio [aHR], 0.84; 95% confidence interval [CI], 0.76-0.94) and had a lower risk of all-cause mortality (aHR, 0.75; 95% CI, 0.66-0.85).

Many disease and patient factors must be considered when choosing the right therapy for a patient. These studies provide valuable information to incorporate into this process and highlight the utility of topical and oral therapies for psoriasis.

Laura Ferris, MD, PhD
While biologics have revolutionized the treatment of psoriasis, oral and topical therapies remain excellent options for many patients. Several recent studies highlight new data on non-biologic therapies.

High-potency topical steroids such as clobetasol are commonly used as first-line treatment for psoriasis. A recent study (Sidgiddi S et al. Dermatol Ther (Heidelb). 2021 Aug 28) compared the efficacy and safety of clobetasol 0.05% vs. 0.025% cream.  Patients were randomized to receive either clobetasol 0.05% cream or one of two different formulations of clobetasol 0.025% cream twice a day for two weeks. PGA success rates (clear or almost clear skin) were higher with the 0.025% formulations (38.9% and 36.8%) than with the 0.05% cream (30.8%). Safety also appeared to be better or comparable with the 0.025% formulation as measured by the proportion of patients with an abnormal ACTH stimulation test (20.7% and 17.2% in the 0.025% group compared with 30.0% in the 0.05% group). Due to the small study size (88 subjects) these differences did not reach statistical significance, although they suggest that high efficacy and perhaps better safety (reduced hypothalamic–pituitary–adrenal axis suppression) can be achieved with lower concentration formulations of clobetasol cream.

The oral phosphodiesterase 4 inhibitor apremilast is FDA-approved to treat psoriasis and psoriatic arthritis and recent studies have shown that it is more effective than placebo in treating patients with mild-moderate psoriasis and scalp psoriasis. A recent prospective cohort study followed 45 adult patients with plaque and nail psoriasis with a fingernail Nail Psoriasis Severity Index (NAPSI) score of 12 or more treated with apremilast 30 mg twice daily for 52 weeks (Muñoz-Santos C et al. J Dermatol. 2021 Aug 12). The primary endpoint, the percentage of patients with a Nail Assessment in Psoriasis and Psoriatic Arthritis-Patient Benefit Index of 2 or more at week 52, was achieved in 52% of patients. A median improvement of 53% in fingernail NAPSI score and a mean reduction in nail pain of 68% were observed at week 52.  These findings show that apremilast can be useful in improving the quality-of-life impairment caused by nail psoriasis.

Traditional systemic therapies such as methotrexate and acitretin are cost-effective options for many psoriasis patients. The association of psoriasis with other comorbid conditions, particularly cardiovascular disease, has raised awareness of the importance of considering how therapies impact not just skin disease but also the risk posed by these psoriasis-associated comorbidities. A large Taiwanese retrospective cohort study compared patients treated with methotrexate (13,777) or acitretin (6,020) and found that in comparison to those treated with acitretin, patients treated with methotrexate were at lower risk of experiencing adverse cardiovascular outcomes, including ischemic heart disease and stroke, (adjusted hazard ratio [aHR], 0.84; 95% confidence interval [CI], 0.76-0.94) and had a lower risk of all-cause mortality (aHR, 0.75; 95% CI, 0.66-0.85).

Many disease and patient factors must be considered when choosing the right therapy for a patient. These studies provide valuable information to incorporate into this process and highlight the utility of topical and oral therapies for psoriasis.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Psoriasis October 2021
Gate On Date
Tue, 08/31/2021 - 12:00
Un-Gate On Date
Tue, 08/31/2021 - 12:00
Use ProPublica
CFC Schedule Remove Status
Tue, 08/31/2021 - 12:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: AML October 2021

Article Type
Changed
Fri, 05/06/2022 - 16:50
Dr. Atallah scans the journals, so you don’t have to!

Ehab Atallah, MD

This month a couple of studies evaluated the role of enasidenib and gemtuzumab in the therapy of patients with AML using clinical and non-clinical trial data. In a propensity score-matching (PSM) analysis, was enasidenib associated with better outcomes compared to standard of care (SoC). The study included adult patients with relapsed/refractory (R/R) AML with an isocitrate dehydrogenase 2 (IDH2) mutation ineligible for hematopoietic stem cell transplantation (HSCT) treated with enasidenib (n = 195) from the AG221 C-001 trial or SoC therapies (n = 78) from the French Chart Review study. The SoC regimens included azacytidine, decitabine, cytarabine containing regimens, and others. Enasidenib was associated with superior overall survival (OS) (hazard ratio [HR] 0.67; 95% CI 0.47-0.97) compared to SoC therapies in patients with R/R AML with IDH2 mutation who were ineligible for HSCT. The median OS was 9.26 months vs. 4.76 months for enasidenib and SoC respectively. The results of this study are not surprising given the data seen in clinical trials. Although not evaluated in this study, the quality of life for patients receiving enasidenib is better when compared to SoC regimens, such as 7 +3 (cytarabine + idarubicin). Other than the limitations of a PSM study, which are well acknowledged by the authors, the main weakness is this study did not include venetoclax in the SoC regimen. It remains unclear whether using venetoclax in the R/R setting in patients with IDH2 mutations would lead to similar outcomes.

 

The benefit of gemtuzumab ozogamicin (GO) was demonstrated again in a metanalysis that included 15 randomized controlled trials and 15 retrospective studies that compared GO (n = 4,768) with non-GO (n = 6,466) therapies in 11,234 patients with AML (n = 11,105) and high-risk myelodysplastic syndrome (MDS) (n = 129). GO showed superior survival outcomes in patients with AML or high-risk MDS than non-GO therapy, but higher doses increased the risk for early death. GO vs. non-GO arm showed improved overall survival (HR 0.86; P = .003), event-free survival (HR 0.86; P = .015), and relapse-free survival (HR 0.83; P = .001). However, GO vs. non-GO was associated with an increased risk for early death at a dose of ≥6 mg/m2 (relative risk [RR] 2.01; P = .005), hepatic-related adverse effects (RR 1.29; P = .02), and bleeding (RR 1.13; P = .018). Finally, a study by the Polish adult leukemia group demonstrated the safety and efficacy of a low dose cytarabine + cladribine regimen. Treatment consisted of 5 mg/m2 cladribine intravenously on day 1-5 (in the first cycle) and on days 1 to 3 (in the second cycle) combined with low dose cytarabine (40 mg) subcutaneously once daily on days 1–10. Patients may continue on low dose cytarabine after that if they have achieved at least a partial remission. Rates of overall response and 56-day mortality were 54.0% and 20.5%, respectively. The median overall survival was 6.9 months (95% CI 4.9-9.6) and superior in patients who achieved an objective response (HR 0.04; P < .0001) or partial remission (HR 0.31; P = .0002) compared to nonresponders.

 

In addition to the better therapies in the last decade, allogeneic hematopoietic stem cell transplantation (allo-HSCT) outcomes have also improved. In a retrospective analysis from the Japanese nationwide transplantation registry the outcomes of patients transplanted with unrelated cord blood (UCB) and Haplo transplant between 2007 to 2014 were worse when compared to patients who had a transplant between 2015 and 2018. For patients who had transplants between 2015 and 2018, the 3-year overall survival of mismatched unrelated donor (MMUD), unrelated cord blood (UCB), and Haplo groups were 60%, 66%, and 63%, respectively (P = .693). The rates of nonrelapse mortality (P = .147), relapse (P = .075), and graft-vs. host disease-free/relapse-free survival (P = .365) were also similar between the 3 groups. For patients with transplant between 2007 to 2014 the 3 year overall survival was 60%, 54%, and 47% for MMUD, UCB, and Haplo transplants respectively. Newer, better drugs, better tolerated regimens, and improved transplant outcomes were the result: many small gains lead to large improvements.

 

 

Author and Disclosure Information

Ehab L. Atallah, MD

Medical College of Wisconsin

Publications
Topics
Sections
Author and Disclosure Information

Ehab L. Atallah, MD

Medical College of Wisconsin

Author and Disclosure Information

Ehab L. Atallah, MD

Medical College of Wisconsin

Dr. Atallah scans the journals, so you don’t have to!
Dr. Atallah scans the journals, so you don’t have to!

Ehab Atallah, MD

This month a couple of studies evaluated the role of enasidenib and gemtuzumab in the therapy of patients with AML using clinical and non-clinical trial data. In a propensity score-matching (PSM) analysis, was enasidenib associated with better outcomes compared to standard of care (SoC). The study included adult patients with relapsed/refractory (R/R) AML with an isocitrate dehydrogenase 2 (IDH2) mutation ineligible for hematopoietic stem cell transplantation (HSCT) treated with enasidenib (n = 195) from the AG221 C-001 trial or SoC therapies (n = 78) from the French Chart Review study. The SoC regimens included azacytidine, decitabine, cytarabine containing regimens, and others. Enasidenib was associated with superior overall survival (OS) (hazard ratio [HR] 0.67; 95% CI 0.47-0.97) compared to SoC therapies in patients with R/R AML with IDH2 mutation who were ineligible for HSCT. The median OS was 9.26 months vs. 4.76 months for enasidenib and SoC respectively. The results of this study are not surprising given the data seen in clinical trials. Although not evaluated in this study, the quality of life for patients receiving enasidenib is better when compared to SoC regimens, such as 7 +3 (cytarabine + idarubicin). Other than the limitations of a PSM study, which are well acknowledged by the authors, the main weakness is this study did not include venetoclax in the SoC regimen. It remains unclear whether using venetoclax in the R/R setting in patients with IDH2 mutations would lead to similar outcomes.

 

The benefit of gemtuzumab ozogamicin (GO) was demonstrated again in a metanalysis that included 15 randomized controlled trials and 15 retrospective studies that compared GO (n = 4,768) with non-GO (n = 6,466) therapies in 11,234 patients with AML (n = 11,105) and high-risk myelodysplastic syndrome (MDS) (n = 129). GO showed superior survival outcomes in patients with AML or high-risk MDS than non-GO therapy, but higher doses increased the risk for early death. GO vs. non-GO arm showed improved overall survival (HR 0.86; P = .003), event-free survival (HR 0.86; P = .015), and relapse-free survival (HR 0.83; P = .001). However, GO vs. non-GO was associated with an increased risk for early death at a dose of ≥6 mg/m2 (relative risk [RR] 2.01; P = .005), hepatic-related adverse effects (RR 1.29; P = .02), and bleeding (RR 1.13; P = .018). Finally, a study by the Polish adult leukemia group demonstrated the safety and efficacy of a low dose cytarabine + cladribine regimen. Treatment consisted of 5 mg/m2 cladribine intravenously on day 1-5 (in the first cycle) and on days 1 to 3 (in the second cycle) combined with low dose cytarabine (40 mg) subcutaneously once daily on days 1–10. Patients may continue on low dose cytarabine after that if they have achieved at least a partial remission. Rates of overall response and 56-day mortality were 54.0% and 20.5%, respectively. The median overall survival was 6.9 months (95% CI 4.9-9.6) and superior in patients who achieved an objective response (HR 0.04; P < .0001) or partial remission (HR 0.31; P = .0002) compared to nonresponders.

 

In addition to the better therapies in the last decade, allogeneic hematopoietic stem cell transplantation (allo-HSCT) outcomes have also improved. In a retrospective analysis from the Japanese nationwide transplantation registry the outcomes of patients transplanted with unrelated cord blood (UCB) and Haplo transplant between 2007 to 2014 were worse when compared to patients who had a transplant between 2015 and 2018. For patients who had transplants between 2015 and 2018, the 3-year overall survival of mismatched unrelated donor (MMUD), unrelated cord blood (UCB), and Haplo groups were 60%, 66%, and 63%, respectively (P = .693). The rates of nonrelapse mortality (P = .147), relapse (P = .075), and graft-vs. host disease-free/relapse-free survival (P = .365) were also similar between the 3 groups. For patients with transplant between 2007 to 2014 the 3 year overall survival was 60%, 54%, and 47% for MMUD, UCB, and Haplo transplants respectively. Newer, better drugs, better tolerated regimens, and improved transplant outcomes were the result: many small gains lead to large improvements.

 

 

Ehab Atallah, MD

This month a couple of studies evaluated the role of enasidenib and gemtuzumab in the therapy of patients with AML using clinical and non-clinical trial data. In a propensity score-matching (PSM) analysis, was enasidenib associated with better outcomes compared to standard of care (SoC). The study included adult patients with relapsed/refractory (R/R) AML with an isocitrate dehydrogenase 2 (IDH2) mutation ineligible for hematopoietic stem cell transplantation (HSCT) treated with enasidenib (n = 195) from the AG221 C-001 trial or SoC therapies (n = 78) from the French Chart Review study. The SoC regimens included azacytidine, decitabine, cytarabine containing regimens, and others. Enasidenib was associated with superior overall survival (OS) (hazard ratio [HR] 0.67; 95% CI 0.47-0.97) compared to SoC therapies in patients with R/R AML with IDH2 mutation who were ineligible for HSCT. The median OS was 9.26 months vs. 4.76 months for enasidenib and SoC respectively. The results of this study are not surprising given the data seen in clinical trials. Although not evaluated in this study, the quality of life for patients receiving enasidenib is better when compared to SoC regimens, such as 7 +3 (cytarabine + idarubicin). Other than the limitations of a PSM study, which are well acknowledged by the authors, the main weakness is this study did not include venetoclax in the SoC regimen. It remains unclear whether using venetoclax in the R/R setting in patients with IDH2 mutations would lead to similar outcomes.

 

The benefit of gemtuzumab ozogamicin (GO) was demonstrated again in a metanalysis that included 15 randomized controlled trials and 15 retrospective studies that compared GO (n = 4,768) with non-GO (n = 6,466) therapies in 11,234 patients with AML (n = 11,105) and high-risk myelodysplastic syndrome (MDS) (n = 129). GO showed superior survival outcomes in patients with AML or high-risk MDS than non-GO therapy, but higher doses increased the risk for early death. GO vs. non-GO arm showed improved overall survival (HR 0.86; P = .003), event-free survival (HR 0.86; P = .015), and relapse-free survival (HR 0.83; P = .001). However, GO vs. non-GO was associated with an increased risk for early death at a dose of ≥6 mg/m2 (relative risk [RR] 2.01; P = .005), hepatic-related adverse effects (RR 1.29; P = .02), and bleeding (RR 1.13; P = .018). Finally, a study by the Polish adult leukemia group demonstrated the safety and efficacy of a low dose cytarabine + cladribine regimen. Treatment consisted of 5 mg/m2 cladribine intravenously on day 1-5 (in the first cycle) and on days 1 to 3 (in the second cycle) combined with low dose cytarabine (40 mg) subcutaneously once daily on days 1–10. Patients may continue on low dose cytarabine after that if they have achieved at least a partial remission. Rates of overall response and 56-day mortality were 54.0% and 20.5%, respectively. The median overall survival was 6.9 months (95% CI 4.9-9.6) and superior in patients who achieved an objective response (HR 0.04; P < .0001) or partial remission (HR 0.31; P = .0002) compared to nonresponders.

 

In addition to the better therapies in the last decade, allogeneic hematopoietic stem cell transplantation (allo-HSCT) outcomes have also improved. In a retrospective analysis from the Japanese nationwide transplantation registry the outcomes of patients transplanted with unrelated cord blood (UCB) and Haplo transplant between 2007 to 2014 were worse when compared to patients who had a transplant between 2015 and 2018. For patients who had transplants between 2015 and 2018, the 3-year overall survival of mismatched unrelated donor (MMUD), unrelated cord blood (UCB), and Haplo groups were 60%, 66%, and 63%, respectively (P = .693). The rates of nonrelapse mortality (P = .147), relapse (P = .075), and graft-vs. host disease-free/relapse-free survival (P = .365) were also similar between the 3 groups. For patients with transplant between 2007 to 2014 the 3 year overall survival was 60%, 54%, and 47% for MMUD, UCB, and Haplo transplants respectively. Newer, better drugs, better tolerated regimens, and improved transplant outcomes were the result: many small gains lead to large improvements.

 

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: AML October 2021
Gate On Date
Fri, 05/07/2021 - 13:30
Un-Gate On Date
Fri, 05/07/2021 - 13:30
Use ProPublica
CFC Schedule Remove Status
Fri, 05/07/2021 - 13:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Clinical Edge Journal Scan Commentary: Prostate Cancer October 2021

Article Type
Changed
Thu, 05/12/2022 - 11:57
Dr. Klein scans the journals, so you don’t have to!

Mark Klein, MD

            Radiation has long been a part of treatment for localized prostate cancer. However, radiation and radiation-delivery modalities continue to evolve to decrease toxicity without sacrificing efficacy, expand the roles of these modalities, and enhance efficacy of other concomitantly delivered treatment modalities. The accompanying studies describe efforts to those effects.

            Hypofractionated delivery of radiation is now often considered preferred over conventionally fractionated radiation for localized prostate cancer to maintain efficacy while decreasing toxicity. To determine if the benefit of lower toxicity holds over time, Staffurth et al assessed patient-reported outcomes (PROs) in participants of the CHHiP trial where men were randomized to conventional radiation versus 2 alternative hypofraction schedules. At 5 years post-radiation, there were no significant differences amongst the 3 groups with respect to bowel bother, urinary bother, or sexual bother.

            Evidence is emerging that stereotactic body radiation therapy (SBRT) has benefits for oligometastatic disease; however, studies are emerging that support the hypothesis that radiation may enhance immunotherapy in many cancers in the metastatic setting. Kwan et al evaluated the effects of a single fraction of SBRT administered to 1 or 2 disease sites before the first and second doses of avelumab (a PD-L1 antibody) in patients with metastatic castrate-resistant prostate cancer (with prior exposure to at least one second-generation androgen receptor inhibitor). In this single arm phase II study, the disease control rate was 48%, overall response rate was 31%, median radiographic progression-free survival (rPFS) was 8.4 months, and median overall survival (OS) was 14.1 months in this heavily pretreated group of 31 participants. As immunotherapy has been considered efficacious in select groups of prostate cancer (such as tumors with microsatellite instability), this study supports ongoing and new efforts to evaluate how to enhance immunotherapy against prostate cancer.

            Radium-223 has been demonstrated to have efficacy in metastatic disease of the bone. Ongoing efforts to determine whether combined modality treatment in this setting may improve outcomes in metastatic disease, Maughn et al evaluated the combination of radium-223 and enzalutamide for safety and efficacy in a small and hypothesis-generating study 47 participants where 35 received enzalutamide plus radium-223 and 12 received enzlutamide alone. Of note, there were no increase in fractures (45 of the 47 participants received bone protective therapy, however), but there were no differences in the secondary endpoints of PSA-PFS, rPFS, or OS (primary endpoints of decline in bone metabolism markers were previously reported). This is a hypothesis generating study that supports the likely safety of this modality if bone protecting agents are utilized.

            The 3 studies summarized here represent the ongoing evolution of radiation or radiation-delivery modalities in localized and metastatic prostate cancer. It is encouraging to see the ongoing evaluate of PROs over time, as long-term quality of life in patients potentially cured of disease is of utmost importance. In addition, efforts to evaluate radiation or radium-223 in widely metastatic disease as part of combination therapy may reveal not only situations with modest benefits in efficacy, but more importantly, may reveal molecular insights into newer treatment strategies.

Author and Disclosure Information

Mark Klein, MD

Minneapolis VA Health Care System

University of Minnesota

Publications
Topics
Sections
Author and Disclosure Information

Mark Klein, MD

Minneapolis VA Health Care System

University of Minnesota

Author and Disclosure Information

Mark Klein, MD

Minneapolis VA Health Care System

University of Minnesota

Dr. Klein scans the journals, so you don’t have to!
Dr. Klein scans the journals, so you don’t have to!

Mark Klein, MD

            Radiation has long been a part of treatment for localized prostate cancer. However, radiation and radiation-delivery modalities continue to evolve to decrease toxicity without sacrificing efficacy, expand the roles of these modalities, and enhance efficacy of other concomitantly delivered treatment modalities. The accompanying studies describe efforts to those effects.

            Hypofractionated delivery of radiation is now often considered preferred over conventionally fractionated radiation for localized prostate cancer to maintain efficacy while decreasing toxicity. To determine if the benefit of lower toxicity holds over time, Staffurth et al assessed patient-reported outcomes (PROs) in participants of the CHHiP trial where men were randomized to conventional radiation versus 2 alternative hypofraction schedules. At 5 years post-radiation, there were no significant differences amongst the 3 groups with respect to bowel bother, urinary bother, or sexual bother.

            Evidence is emerging that stereotactic body radiation therapy (SBRT) has benefits for oligometastatic disease; however, studies are emerging that support the hypothesis that radiation may enhance immunotherapy in many cancers in the metastatic setting. Kwan et al evaluated the effects of a single fraction of SBRT administered to 1 or 2 disease sites before the first and second doses of avelumab (a PD-L1 antibody) in patients with metastatic castrate-resistant prostate cancer (with prior exposure to at least one second-generation androgen receptor inhibitor). In this single arm phase II study, the disease control rate was 48%, overall response rate was 31%, median radiographic progression-free survival (rPFS) was 8.4 months, and median overall survival (OS) was 14.1 months in this heavily pretreated group of 31 participants. As immunotherapy has been considered efficacious in select groups of prostate cancer (such as tumors with microsatellite instability), this study supports ongoing and new efforts to evaluate how to enhance immunotherapy against prostate cancer.

            Radium-223 has been demonstrated to have efficacy in metastatic disease of the bone. Ongoing efforts to determine whether combined modality treatment in this setting may improve outcomes in metastatic disease, Maughn et al evaluated the combination of radium-223 and enzalutamide for safety and efficacy in a small and hypothesis-generating study 47 participants where 35 received enzalutamide plus radium-223 and 12 received enzlutamide alone. Of note, there were no increase in fractures (45 of the 47 participants received bone protective therapy, however), but there were no differences in the secondary endpoints of PSA-PFS, rPFS, or OS (primary endpoints of decline in bone metabolism markers were previously reported). This is a hypothesis generating study that supports the likely safety of this modality if bone protecting agents are utilized.

            The 3 studies summarized here represent the ongoing evolution of radiation or radiation-delivery modalities in localized and metastatic prostate cancer. It is encouraging to see the ongoing evaluate of PROs over time, as long-term quality of life in patients potentially cured of disease is of utmost importance. In addition, efforts to evaluate radiation or radium-223 in widely metastatic disease as part of combination therapy may reveal not only situations with modest benefits in efficacy, but more importantly, may reveal molecular insights into newer treatment strategies.

Mark Klein, MD

            Radiation has long been a part of treatment for localized prostate cancer. However, radiation and radiation-delivery modalities continue to evolve to decrease toxicity without sacrificing efficacy, expand the roles of these modalities, and enhance efficacy of other concomitantly delivered treatment modalities. The accompanying studies describe efforts to those effects.

            Hypofractionated delivery of radiation is now often considered preferred over conventionally fractionated radiation for localized prostate cancer to maintain efficacy while decreasing toxicity. To determine if the benefit of lower toxicity holds over time, Staffurth et al assessed patient-reported outcomes (PROs) in participants of the CHHiP trial where men were randomized to conventional radiation versus 2 alternative hypofraction schedules. At 5 years post-radiation, there were no significant differences amongst the 3 groups with respect to bowel bother, urinary bother, or sexual bother.

            Evidence is emerging that stereotactic body radiation therapy (SBRT) has benefits for oligometastatic disease; however, studies are emerging that support the hypothesis that radiation may enhance immunotherapy in many cancers in the metastatic setting. Kwan et al evaluated the effects of a single fraction of SBRT administered to 1 or 2 disease sites before the first and second doses of avelumab (a PD-L1 antibody) in patients with metastatic castrate-resistant prostate cancer (with prior exposure to at least one second-generation androgen receptor inhibitor). In this single arm phase II study, the disease control rate was 48%, overall response rate was 31%, median radiographic progression-free survival (rPFS) was 8.4 months, and median overall survival (OS) was 14.1 months in this heavily pretreated group of 31 participants. As immunotherapy has been considered efficacious in select groups of prostate cancer (such as tumors with microsatellite instability), this study supports ongoing and new efforts to evaluate how to enhance immunotherapy against prostate cancer.

            Radium-223 has been demonstrated to have efficacy in metastatic disease of the bone. Ongoing efforts to determine whether combined modality treatment in this setting may improve outcomes in metastatic disease, Maughn et al evaluated the combination of radium-223 and enzalutamide for safety and efficacy in a small and hypothesis-generating study 47 participants where 35 received enzalutamide plus radium-223 and 12 received enzlutamide alone. Of note, there were no increase in fractures (45 of the 47 participants received bone protective therapy, however), but there were no differences in the secondary endpoints of PSA-PFS, rPFS, or OS (primary endpoints of decline in bone metabolism markers were previously reported). This is a hypothesis generating study that supports the likely safety of this modality if bone protecting agents are utilized.

            The 3 studies summarized here represent the ongoing evolution of radiation or radiation-delivery modalities in localized and metastatic prostate cancer. It is encouraging to see the ongoing evaluate of PROs over time, as long-term quality of life in patients potentially cured of disease is of utmost importance. In addition, efforts to evaluate radiation or radium-223 in widely metastatic disease as part of combination therapy may reveal not only situations with modest benefits in efficacy, but more importantly, may reveal molecular insights into newer treatment strategies.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Prostate Cancer October 2021
Gate On Date
Tue, 06/01/2021 - 10:45
Un-Gate On Date
Tue, 06/01/2021 - 10:45
Use ProPublica
CFC Schedule Remove Status
Tue, 06/01/2021 - 10:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
sf322941.4
Activity ID
77695
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
xtandi [ 4408 ]

Clinical Edge Journal Scan Commentary: HCC October 2021

Article Type
Changed
Thu, 05/12/2022 - 11:57
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD

Treatment of patients with hepatocellular carcinoma (HCC) requires a multidisciplinary approach. This month we will review articles that analyze outcomes after liver resection compared to percutaneous ablation, prediction of immunotherapy efficacy based on observed treatment-related side effects, and the risk of subsequent malignancies in patients with HCC.

Xie et al. reported their retrospective review of outcomes of 67 adults with resectable caudate HCC within Milan criteria. Out of these, 46 underwent hepatic resection and 21 underwent percutaneous ablation. Overall survival at 1, 3, and 5 years was 97.6%, 83.6%, and 71.5%, respectively, for the hepatic resection patients, vs 89.4%, 58.5%, and 48.8%, respectively, for the percutaneous ablation patients (P = 0.032). Recurrence-free survival at these time points was 77.6%, 47.9%, and 42.6%, respectively, for the hepatic resection group, and 40.5%, 23.2%, and 15.4%, respectively, for the percutaneous ablation group (P = 0.010). The investigators concluded that HCC patients who underwent hepatic resection had significantly higher rates of recurrence-free and overall survival compared to those who underwent percutaneous ablation.

For those patients with unresectable HCC, systemic immune checkpoint inhibitor therapy is now part of the standard of care. Treatment-associated adverse events occur in over half of patients treated with immunotherapy. Pinato et al. reviewed the outcomes of a cohort of 406 adults with unresectable or advanced HCC who were receiving immune checkpoint inhibitor therapy while enrolled in clinical trials that were submitted to the Food and Drug Administration. the development of adverse events was associated with longer overall survival and progression-free survival rates compared to patients who did not develop treatment-related adverse events (16.7 months vs 11.2 months; 5.5 months vs 2.2 months, respectively).  The authors concluded that the development of treatment-related adverse events was significantly correlated with improve overall and progression-free survival in HCC patients treated with ICI monotherapy in clinical trials.

Finally, Kong et al. retrospectively looked at a cohort of 40,314 adult patients diagnosed with HCC between 2000 and 2014 in the SEER database, identifying the incidence of second primary cancers. Overall, the patients were followed for a median of 19 months following their HCC diagnosis. A total of 1,593 HCC patients (3.95%) developed secondary primary malignancies starting at 2 months after their initial HCC diagnosis. The 3-, 5-, and 10-year cumulative incidence of developing second primary malignancies were 2.35%, 3.12%, and 4.51%. The top five sites of the second primary malignancies were lung and bronchus, prostate, non-Hodgkin lymphoma, colon, and breast. The patients with poorer tumor-related characteristics such as larger tumor size, vascular invasion, positive AFP level, poorer tumor grade, and distant extension were associated with a decreased risk of developing second primary cancers, most probably because of their higher risk of dying from HCC. The authors developed a competing-risk nomogram for the purpose of improving guideline surveillance and further management of HCC survivors, concluding that HCC survivors should be monitored for evidence of secondary primary cancers.

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology,  Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Publications
Topics
Sections
Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology,  Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology,  Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Dr. Damjanov scans the journals, so you don’t have to!
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD

Treatment of patients with hepatocellular carcinoma (HCC) requires a multidisciplinary approach. This month we will review articles that analyze outcomes after liver resection compared to percutaneous ablation, prediction of immunotherapy efficacy based on observed treatment-related side effects, and the risk of subsequent malignancies in patients with HCC.

Xie et al. reported their retrospective review of outcomes of 67 adults with resectable caudate HCC within Milan criteria. Out of these, 46 underwent hepatic resection and 21 underwent percutaneous ablation. Overall survival at 1, 3, and 5 years was 97.6%, 83.6%, and 71.5%, respectively, for the hepatic resection patients, vs 89.4%, 58.5%, and 48.8%, respectively, for the percutaneous ablation patients (P = 0.032). Recurrence-free survival at these time points was 77.6%, 47.9%, and 42.6%, respectively, for the hepatic resection group, and 40.5%, 23.2%, and 15.4%, respectively, for the percutaneous ablation group (P = 0.010). The investigators concluded that HCC patients who underwent hepatic resection had significantly higher rates of recurrence-free and overall survival compared to those who underwent percutaneous ablation.

For those patients with unresectable HCC, systemic immune checkpoint inhibitor therapy is now part of the standard of care. Treatment-associated adverse events occur in over half of patients treated with immunotherapy. Pinato et al. reviewed the outcomes of a cohort of 406 adults with unresectable or advanced HCC who were receiving immune checkpoint inhibitor therapy while enrolled in clinical trials that were submitted to the Food and Drug Administration. the development of adverse events was associated with longer overall survival and progression-free survival rates compared to patients who did not develop treatment-related adverse events (16.7 months vs 11.2 months; 5.5 months vs 2.2 months, respectively).  The authors concluded that the development of treatment-related adverse events was significantly correlated with improve overall and progression-free survival in HCC patients treated with ICI monotherapy in clinical trials.

Finally, Kong et al. retrospectively looked at a cohort of 40,314 adult patients diagnosed with HCC between 2000 and 2014 in the SEER database, identifying the incidence of second primary cancers. Overall, the patients were followed for a median of 19 months following their HCC diagnosis. A total of 1,593 HCC patients (3.95%) developed secondary primary malignancies starting at 2 months after their initial HCC diagnosis. The 3-, 5-, and 10-year cumulative incidence of developing second primary malignancies were 2.35%, 3.12%, and 4.51%. The top five sites of the second primary malignancies were lung and bronchus, prostate, non-Hodgkin lymphoma, colon, and breast. The patients with poorer tumor-related characteristics such as larger tumor size, vascular invasion, positive AFP level, poorer tumor grade, and distant extension were associated with a decreased risk of developing second primary cancers, most probably because of their higher risk of dying from HCC. The authors developed a competing-risk nomogram for the purpose of improving guideline surveillance and further management of HCC survivors, concluding that HCC survivors should be monitored for evidence of secondary primary cancers.

Nevena Damjanov, MD

Treatment of patients with hepatocellular carcinoma (HCC) requires a multidisciplinary approach. This month we will review articles that analyze outcomes after liver resection compared to percutaneous ablation, prediction of immunotherapy efficacy based on observed treatment-related side effects, and the risk of subsequent malignancies in patients with HCC.

Xie et al. reported their retrospective review of outcomes of 67 adults with resectable caudate HCC within Milan criteria. Out of these, 46 underwent hepatic resection and 21 underwent percutaneous ablation. Overall survival at 1, 3, and 5 years was 97.6%, 83.6%, and 71.5%, respectively, for the hepatic resection patients, vs 89.4%, 58.5%, and 48.8%, respectively, for the percutaneous ablation patients (P = 0.032). Recurrence-free survival at these time points was 77.6%, 47.9%, and 42.6%, respectively, for the hepatic resection group, and 40.5%, 23.2%, and 15.4%, respectively, for the percutaneous ablation group (P = 0.010). The investigators concluded that HCC patients who underwent hepatic resection had significantly higher rates of recurrence-free and overall survival compared to those who underwent percutaneous ablation.

For those patients with unresectable HCC, systemic immune checkpoint inhibitor therapy is now part of the standard of care. Treatment-associated adverse events occur in over half of patients treated with immunotherapy. Pinato et al. reviewed the outcomes of a cohort of 406 adults with unresectable or advanced HCC who were receiving immune checkpoint inhibitor therapy while enrolled in clinical trials that were submitted to the Food and Drug Administration. the development of adverse events was associated with longer overall survival and progression-free survival rates compared to patients who did not develop treatment-related adverse events (16.7 months vs 11.2 months; 5.5 months vs 2.2 months, respectively).  The authors concluded that the development of treatment-related adverse events was significantly correlated with improve overall and progression-free survival in HCC patients treated with ICI monotherapy in clinical trials.

Finally, Kong et al. retrospectively looked at a cohort of 40,314 adult patients diagnosed with HCC between 2000 and 2014 in the SEER database, identifying the incidence of second primary cancers. Overall, the patients were followed for a median of 19 months following their HCC diagnosis. A total of 1,593 HCC patients (3.95%) developed secondary primary malignancies starting at 2 months after their initial HCC diagnosis. The 3-, 5-, and 10-year cumulative incidence of developing second primary malignancies were 2.35%, 3.12%, and 4.51%. The top five sites of the second primary malignancies were lung and bronchus, prostate, non-Hodgkin lymphoma, colon, and breast. The patients with poorer tumor-related characteristics such as larger tumor size, vascular invasion, positive AFP level, poorer tumor grade, and distant extension were associated with a decreased risk of developing second primary cancers, most probably because of their higher risk of dying from HCC. The authors developed a competing-risk nomogram for the purpose of improving guideline surveillance and further management of HCC survivors, concluding that HCC survivors should be monitored for evidence of secondary primary cancers.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: HCC October 2021
Gate On Date
Mon, 05/17/2021 - 15:00
Un-Gate On Date
Mon, 05/17/2021 - 15:00
Use ProPublica
CFC Schedule Remove Status
Mon, 05/17/2021 - 15:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article