The FDA on July 20, 2023, approved VANFLYTA® (Quizartinib) with standard Cytarabine and Anthracycline induction and Cytarabine consolidation, and as maintenance monotherapy following consolidation chemotherapy, for the treatment of adult patients with newly diagnosed Acute Myeloid Leukemia (AML) that is FLT3 Internal Tandem Duplication (ITD)-positive, as detected by an FDA-approved test. VANFLYTA® is a product of Daiichi Sankyo, Inc.
Author: RR
TALZENNA® (Talazoparib) with Enzalutamide
The FDA on June 20, 2023, approved TALZENNA® with Enzalutamide for Homologous Recombination Repair (HRR) gene-mutated metastatic Castration-Resistant Prostate Cancer (mCRPC). TALZENNA® is a product of Pfizer, Inc.
COLUMVI® (Glofitamab-gxbm)
The FDA on June 15, 2023, granted accelerated approval to COLUMVI® for Relapsed or Refractory Diffuse Large B-Cell Lymphoma, not otherwise specified (DLBCL, NOS) or Large B-Cell Lymphoma (LBCL) arising from Follicular Lymphoma, after two or more lines of systemic therapy. COLUMVI® is a product of Genentech, Inc.
LYNPARZA® (Olaparib)
The FDA on May 31, 2023, approved LYNPARZA® (Olaparib) along with Abiraterone and Prednisone (or Prednisolone) for adult patients with deleterious or suspected deleterious BRCA-mutated (BRCAm) metastatic Castration-Resistant Prostate Cancer (mCRPC), as determined by an FDA-approved companion diagnostic test. LYNPARZA® is a product of AstraZeneca Pharmaceuticals LP.
EPKINLY® (Epcoritamab-bysp)
The FDA on May 19, 2023 granted accelerated approval to EPKINLY® for Relapsed or Refractory Diffuse Large B-Cell Lymphoma (DLBCL) not otherwise specified, including DLBCL arising from indolent lymphoma, and high-grade B-cell lymphoma after two or more lines of systemic therapy. EPKINLY® is a product of Genmab US, Inc.
POLIVY® (Polatuzumab vedotin-piiq)
The FDA on April 19, 2023, approved POLIVY® with a Rituximab product, Cyclophosphamide, Doxorubicin, and Prednisone (R-CHP) for adult patients who have previously untreated Diffuse Large B-Cell Lymphoma (DLBCL), not otherwise specified (NOS), or High-Grade B-Cell Lymphoma (HGBL) and who have an International Prognostic Index (IPI) score of 2 or greater. POLIVY® is a product of Genentech, Inc.
Single Blood Test for Multi-Cancer Early Detection
SUMMARY: The American Cancer Society estimates that in 2023, 1,958,310 new cancer cases and 609,820 cancer deaths are projected to occur in the United States. Although cancer mortality rates continue to decline with advances in treatment, improving early detection can reduce disease and treatment-related morbidity, improve treatment outcomes, quality of life and reduce financial burden both for the patient as well as the society as a whole. Currently the USPSTF (Unites States Preventive Services Task Force) and ACS (American Cancer Society) recommend screening for breast, cervical, colorectal, and lung cancers. Neither the ACS nor USPSTF have specific recommendations for prostate cancer screening. These cancers collectively account for only 42% of annual cancer incidence in people aged 50-79 years. It has been estimated that detection of cancer at an earlier stage could reduce cancer-related deaths by 15% or more within 5 years. Some of the available screening tests reduce cancer-specific mortality, but are associated with high false-positive rates, overdiagnosis, and overtreatment.
Galleri is a Multi-Cancer Early Detection (MCED) test developed for the early detection of multiple asymptomatic cancers that lack recommended screening tests, using a blood sample. DNA (cell free DNA) is shed into the blood stream both by tumor cells as well as healthy cells. The Galleri test uses Next Generation Sequencing (NGS) and machine-learning algorithms to isolate cell-free DNA and analyze more than 100,000 DNA regions and over a million specific DNA sites, to screen for a signal shared by cancers. The test looks for cell-free DNA and identifies whether it comes from healthy or cancer cells. DNA from cancer cells has specific methylation patterns that identify it as a cancer signal. Methylation patterns also contain information about the tissue type or organ associated with the cancer signal. So, once a cancer signal is detected, the Galleri test predicts the Cancer Signal Origin, or the tissue or organ where the cancer signal originated, to help guide diagnostic evaluation. The Galleri test is recommended for use in adults with an elevated risk for cancer, such as those aged 50 or older, and should be used in addition to routine cancer screening tests. Galleri is not recommended in individuals who are pregnant, 21 years old or younger, or undergoing active cancer treatment.
A validation study (Circulating Cell-free Genome Atlas study-CCGA) was conducted to evaluate the accuracy of the Galleri test. This study included 2,823 people with a known diagnosis of cancer and 1,254 healthy people. The overall Sensitivity for cancer signal detection was 51.5% and the Specificity was 99.5%. The sensitivity of the test increased with advanced cancer stages. Cancer signals were detected across over 50 cancer types and the overall accuracy of predicting Cancer Signal Origin in those who tested true positive was 88.7% (Ann Oncol. 2021;32:1167-1177).
PATHFINDER was a pilot, prospective cohort study conducted to investigate the feasibility of MCED testing for cancer screening. This study included 6,621 participants from oncology and primary care outpatient clinics at seven U.S. health networks who underwent MCED blood testing. Participants were 50 years or older, with no signs or symptoms of cancer, and majority were women (63.5%) and White (91.7%). Approximately 56% of participants had additional risk factors such as smoking, germline cancer predisposition, or personal history of treated cancer. The Primary outcome was time to diagnosis, and extent of diagnostic testing required to confirm the presence or absence of cancer.
MCED testing detected a cancer signal in 1.4% of the total patient sample of whom 38% had cancer confirmed (true positives), while 62% had no cancer (false positives). In patients in whom no cancer signal was detected, 95.5% were true negatives, 1.3% was false negatives, and 3.2% did not have cancer-status assessment at the end of the study. The tests accuracy in predicting the primary cancer location (Cancer Signal Origin) among the true positives was high at 97%. The median time to achieving a diagnostic resolution was 79 days, 57 days in true-positive patients and 162 days in false-positive ones. Fewer procedures were done in participants with false-positive results compared to true-positive results (30% versus 82% respectively) and few participants had surgery (one with a false-positive result and three with a true-positive result).
Among participants whose testing was true-positive and who had a confirmed new cancer diagnosis, nearly half (48%) were detected at an early stage (Stage I-II) when the potential for curative treatment is increased. Further, 74% of the MCED-detected cancers were cancer types that do not currently have USPSTF screening recommendations. These included cancers of the bile duct, pancreas, small intestine, and spindle cell neoplasm, which are all associated with high mortality rates and may be amenable to surgical resection at early stages.
In the 12 months study period, 121 cancers were diagnosed, of whom 29% had a cancer signal detected by MCED, while 31% were detected thru screening and 40% were detected clinically. The overall Positive Predictive Value of MCED was 38%, Negative Predictive Value was 98.6%, and specificity was 99.1%. The cancer yield rate was 0.53% (number needed to screen to find one MCED-detected cancer was 189).
The researchers concluded that this study demonstrates the feasibility of screening for multiple cancers using a blood test and lays the foundation for large, controlled trials necessary to establish clinical utility and cost-effectiveness. Multi Cancer Early Detection test was also able to accurately predict tumor origin, and the diagnosis of cancer was established in less than 2 months in the true-positive patients.
Blood-based tests for multicancer early detection (PATHFINDER): a prospective cohort study. Schrag D, Beer TM, McDonnell CH, et al. The Lancet 2023;402:1251-1260.
KEYTRUDA® in Combination with HER2 Blockade Improves PFS in Gastric and GE Junction Cancer
SUMMARY: Gastroesophageal cancers consist of a group of heterogeneous tumors, including gastric cancer, gastroesophageal junction cancer, and esophageal cancer. The majority of gastric and gastroesophageal junction cancers are adenocarcinomas, while the two main histological subtypes of esophageal cancer are esophageal adenocarcinoma and esophageal squamous cell carcinoma. The American Cancer Society estimates that in the US about 26,500 new gastric cancer cases and 21,560 new esophageal cancers will be diagnosed in 2023 and about 11,130 and 16,120 people respectively, will die of the disease. It is one of the leading causes of cancer-related deaths in the world. Patients with localized disease (Stage II and Stage III) are often treated with multimodality therapy and 40% of the patients may survive for 5 years or more. However, majority of the patients with gastric and gastroesophageal junction adenocarcinoma have advanced disease at the time of initial presentation and have limited therapeutic options with little or no chance for cure.
The Human Epidermal growth factor Receptor (HER) or erbB family of receptors, consist of HER1, HER2, HER3 and HER4. Approximately 15-20% of advanced gastric and gastroesophageal (GE) junction cancers, overexpress or have amplification of the HER2 oncogene. These patients often receive first line treatment with a combination of chemotherapy plus anti-HER2 antibody, Trastuzumab, as there is Overall Survival (OS) benefit with this combination regimen. Pembrolizumab (KEYTRUDA®) is a fully humanized, Immunoglobulin G4, anti-PD-1, monoclonal antibody, that binds to the PD-1 receptor expressed on activated T cells, and blocks its interaction with ligands PD-L1 and PD-L2. It thereby reverses the PD-1 pathway-mediated inhibition of the immune response and unleashes the tumor-specific effector T cells. In two Phase II studies, Pembrolizumab in combination with Trastuzumab and chemotherapy showed promising efficacy with manageable toxicities. The FDA in 2021 granted accelerated approval to Pembrolizumab in combination with Trastuzumab, Fluoropyrimidine and Platinum-containing chemotherapy for the first-line treatment of patients with locally advanced unresectable or metastatic HER2 positive gastric or gastroesophageal junction adenocarcinoma, based on Overall Response Rates (ORR).
KEYNOTE-811 is an ongoing, global, multicenter, randomized Phase III trial which evaluated the benefit of adding Pembrolizumab to Trastuzumab and chemotherapy in patients with locally advanced unresectable or metastatic HER2-positive gastric or gastroesophageal junction adenocarcinoma. In this study, 698 treatment naïve eligible patients (N=698) were randomly assigned 1:1 to receive Pembrolizumab 200 mg IV (N=350) or placebo (N=348) every 3 weeks plus Trastuzumab and investigator’s choice of Fluorouracil/Cisplatin or Capecitabine/Oxaliplatin. Trastuzumab was given at 6 mg/kg IV once every 3 weeks after a loading dose of 8 mg/kg IV. Chemotherapy consisted of 5-FU 800 mg/m2 IV on days 1 to 5 of each 3-week cycle and Cisplatin 80 mg/m2 IV once every 3 weeks, or Capecitabine 1,000 mg/m2 orally twice daily on days 1 to 14 of each 3-week cycle and Oxaliplatin 130 mg/m2 IV once every 3 weeks. Treatment was continued for up to 35 cycles or until disease progression or unacceptable toxicity. Approximately 81% were male and patients were stratified by PD-L1 status, and chemotherapy received. Over 80% of patients had a PD-L1 Combined Positive Score of 1 or more. The dual Primary end points of the trial were Progression Free Survival (PFS) and Overall Survival (OS). Secondary end points included Objective Response Rate (ORR), Duration of Response, and Safety. The researchers reported the results at the third interim analysis, after a median follow up of 38.5 months.
At the third interim analysis, the results continued to show superiority with the addition of Pembrolizumab to Trastuzumab and chemotherapy. The median PFS with Pembrolizumab versus placebo was 10 months versus 8.1 months, respectively (HR = 0.73; P=0.0002). This represented a 27% reduction in risk for progression with Pembrolizumab versus placebo. The median OS showed numerical improvement and was 20.0 months versus 16.8 months (HR=0.84), but did not meet prespecified criteria for significance. Follow up for Overall Survival is continuing, and results will be updated at the final analysis. Patients whose tumors had PD-L1 Combined Positive Score of 1 or more benefitted the most, and there was little to no benefit among patients whose tumors had PD-L1 Combined Positive Scores less than 1. The researchers had previously reported an ORR of 74% in the the Pembrolizumab group and 52% in the placebo group, yielding a 22% improvement for the Pembrolizumab group (P=0.00006). Disease Control Rates were 96.2% versus 89.3% respectively. Grade 3 or more treatment-related adverse events were higher among patients assigned to Pembrolizumab versus placebo group (58% versus 51%). The most common treatment-related adverse events of any grade were diarrhea, nausea and anemia.
The authors concluded that Pembrolizumab when combined with first line Trastuzumab and chemotherapy significantly improved Progression Free Survival when compared to placebo, in metastatic HER2-positive gastroesophageal cancer. This benefit was specifically noted among patients with tumors with a PD-L1 Combined Positive Score of 1 or more. Follow up for Overall Survival is ongoing and will be updated at the final analysis.
Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial. Janjigian YY, Kawazoe A, Bai Y, et al. Published:October 20, 2023. DOI:https://doi.org/10.1016/S0140-6736(23)02033-0
FDA Approves Perioperative KEYTRUDA® for Resectable Early Stage Non Small Cell Lung Cancer
SUMMARY: The FDA on October 16, 2023, approved KEYTRUDA® (Pembrolizumab) with platinum-containing chemotherapy as neoadjuvant treatment, and with continuation of single-agent KEYTRUDA® as post-surgical adjuvant treatment for resectable (tumors 4 cm or more or node positive) Non-Small Cell Lung Cancer (NSCLC). Lung cancer is the second most common cancer in both men and women and accounts for about 13% of all new cancers and 21% of all cancer deaths. The American Cancer Society estimates that for 2023, about 238,340 new cases of lung cancer will be diagnosed and 127,070 patients will die of the disease. Lung cancer is the leading cause of cancer-related mortality in the United States. Non-Small Cell Lung Cancer (NSCLC) accounts for approximately 85% of all lung cancers. Of the three main subtypes of NSCLC, 30% are Squamous Cell Carcinomas (SCC), 40% are Adenocarcinomas and 10% are Large Cell Carcinomas. With changes in the cigarette composition and decline in tobacco consumption over the past several decades, Adenocarcinoma now is the most frequent histologic subtype of lung cancer.
The 5-year survival rate for patients diagnosed with lung cancer in the US is about 25%, which is a significant improvement over the past 5 years, in part due to earlier detection from lung cancer screening, reduction in smoking, advances in diagnostic and surgical procedures, as well as the introduction of new therapies. However, the 5-year survival rate remains significantly lower among communities of color at 20%. Early detection and screening remain an important unmet need, as 44% of lung cancer cases are not found until they are advanced. In the US, only 5.8% of those individuals at high risk were screened in 2021.
Surgical resection is the primary treatment for approximately 30% of patients with NSCLC who present with early Stage (I–IIIA) disease. These patients are often treated with platinum-based adjuvant chemotherapy to decrease the risk of recurrence. Nonetheless, 45-75% of these patients develop recurrent disease. There is therefore an unmet need for this patient population.
Immunotherapy with Immune Checkpoint Inhibitors (ICIs) has revolutionized cancer care and has become one of the most effective treatment options, by improving Overall Response Rate and prolongation of survival, across multiple tumor types. These agents target Programmed cell Death protein-1 (PD-1), Programmed cell Death Ligand-1 (PD-L1), Cytotoxic T-Lymphocyte-Associated protein-4 (CTLA-4), and many other important regulators of the immune system. Checkpoint inhibitors unleash the T cells resulting in T cell proliferation, activation, and a therapeutic response. Biomarkers predicting responses to ICIs include Tumor Mutational Burden (TMB), Mismatch Repair (MMR) status, and Programmed cell Death Ligand 1 (PD‐L1) expression. KEYTRUDA® (Pembrolizumab) is a fully humanized, Immunoglobulin G4, anti-PD-1, monoclonal antibody, that binds to the PD-1 receptor and blocks its interaction with ligands PD-L1 and PD-L2.
The present FDA approval was based on the KEYNOTE-671 trial, which is a randomized, double-blind, placebo-controlled, Phase III trial, conducted to evaluate whether a perioperative approach of combined neoadjuvant Pembrolizumab plus Cisplatin-based chemotherapy, followed by surgical resection and adjuvant Pembrolizumab therapy, would improve efficacy as compared with neoadjuvant Cisplatin-based chemotherapy and resection alone, in patients with resectable Stage II or III NSCLC. This study included patients with pathologically confirmed, resectable Stage II, IIIA, or IIIB (N2 disease-with involvement of 1 or more ipsilateral mediastinal lymph nodes or subcarinal lymph node) NSCLC. Eligible patients were randomly assigned in a 1:1 ratio to receive neoadjuvant Pembrolizumab 200 mg IV (N=397) or placebo (N=400) once every 3 weeks, each of which was given with Cisplatin-based chemotherapy for 4 cycles, followed by surgery and adjuvant Pembrolizumab 200 mg IV or placebo once every 3 weeks for up to 13 cycles. The median age was 64 years, 70% had Stage III disease, about 44% had N2 nodal stage, 57% has nonsquamous histology and 43% had squamous histology, about 36% had less than 1% PD-L1 Tumor Proportion Score (TPS), whereas 30% of patients had tumors with a TPS of 1-49% and 33% had TPS of 50% or more. The dual Primary end points were Event-Free Survival (the time from randomization to the first occurrence of local progression that precluded the planned surgery, unresectable tumor, progression or recurrence, or death), and Overall Survival (OS). Secondary end points included major pathological response, pathological Complete Response, and Safety.
The researchers reported the efficacy and safety data from the prespecified first interim analysis. The median follow-up was 25.2 months. The Event-Free Survival (EFS) at 24 months was 62.4% in the Pembrolizumab group and 40.6% in the placebo group. The median EFS was not reached in the Pembrolizumab group and was 17.0 months in the placebo group (HR=0.58; P<0.001). The EFS benefit with Pembrolizumab was consistent across all subgroups examined. The estimated 24-month Overall Survival was 80.9% in the Pembrolizumab group and 77.6% in the placebo group and this was not statistically significant (P=0.02) at this first interim analysis.
A major pathological response occurred in 30.2% of the patients in the Pembrolizumab group and in 11.0% of those in the placebo group (P<0.0001) and a pathological Complete Response occurred in 18.1% and 4.0%, respectively (P<0.0001). An exploratory analysis showed that the Event-Free Survival benefit was noted in the Pembrolizumab group regardless of whether participants had a major pathological response or a pathological Complete Response. The benefit with Pembrolizumab therapy appeared to be similar across both squamous and nonsquamous histologies. Approximately 45% of the patients in the Pembrolizumab group and 37% in the placebo group had treatment-related adverse events of Grade 3 or higher.
It was concluded that among patients with resectable Stage II, IIIA, or IIIB (N2 stage) NSCLC, the addition of Pembrolizumab to neoadjuvant Cisplatin-based chemotherapy, followed by surgical resection and adjuvant Pembrolizumab therapy, led to a significant improvement in Event-Free Survival, major pathological response, and pathological Complete Response, as compared with neoadjuvant chemotherapy alone followed by surgery. It should be noted that this trial was not designed to assess the relative contribution of adjuvant Pembrolizumab.
Perioperative Pembrolizumab for Early-Stage Non–Small-Cell Lung Cancer. Wakelee H, Liberman M, Kato T, et al., for the KEYNOTE-671 Investigators. N Engl J Med 2023;389:491-503.
Hair Relaxer Use and Risk of Uterine Cancer
SUMMARY: The American Cancer Society estimates that approximately 66,200 new cases of uterine cancer will be diagnosed in 2023 and about 13,030 will die of the disease. Uterine cancer is the second most prevalent gynecologic cancer in women worldwide, and its incidence has been increasing. Risk factors include age, factors that influence hormone levels such as obesity and estrogen replacement therapy, Type 2 diabetes, family history, diet and exercise, drugs such as Tamoxifen, and delayed menopause. Exposure to excess estrogen and a hormonal imbalance of estrogen and progesterone have been identified as key risk factors for uterine cancer. Further, synthetic estrogenic compounds such as endocrine-disrupting chemicals have been implicated, because of their ability to alter hormonal actions.
It is estimated that up to 95% of self-identified adult African American women in the U.S. report ever use of hair relaxers. Chemical straighteners or relaxers are commonly used by Black women to straighten curly or tightly coiled hair mostly due to societal pressure to straighten hair. As a result, chemical hair relaxers are heavily marketed to Black women. These straighteners have been found to have potentially harmful toxicants such as phthalates, parabens, cyclosiloxanes and metals that may act as carcinogens or endocrine disruptors, and may release formaldehyde when heated. These products when left on the hair longer than advised can injure the scalp, making it easier to absorb the chemicals. High concentrations of metabolites of phthalates and parabens have been detected in urine samples from women who use chemical hair relaxers.
Endocrine disrupting chemicals may cause hormonal imbalance through either estrogenic or anti-estrogenic activity and has been associated with earlier puberty, infertility, and uterine fibroids. A study published in the International Journal of Cancer in 2020 found that, women who used chemical hair straighteners more than 6 times a year, had about a 30% higher risk of breast cancer. In a study published in the journal Carcinogenesis in 2021 and in the Journal of the National Cancer Institute in 2022 (The Sister Study), women who used chemical hair straighteners more than 4 times a year, were twice as likely to develop ovarian cancer and more than twice as likely to develop uterine cancer as compared to women who did not use chemical hair straighteners. However in the Sister Study there were only 17 exposed cases among Black women. The researchers in this study sought to evaluate the possible association of hair relaxer use with uterine cancer risk in a large prospective cohort of Black women.
The US-based Black Women’s Health Study (BWHS) included 44,798 Black women with an intact uterus and no prior history of cancer, between the ages of 21 and 69, who were followed from 1997 until 2019. All participants completed a self-administered questionnaire about their personal and family medical history as well as behavioral and other factors such as medication use, cigarette smoking, and diet. The researchers used adjusted multivariable Cox proportional hazards regression models, to estimate Hazard Ratios (HRs) and 95% Confidence Intervals (CIs) for associations of hair relaxer use with risk of uterine cancer. The participants were followed for up to 22 years, and the rates of uterine cancer among women who reported frequent or long-term use of hair relaxers were compared to rates among women who never or rarely used them.
It was noted that compared to women who never used hair relaxers or used them infrequently (less than 4 years and 1-2 times or less/year), the Hazard Ratio for uterine cancer associated with heavy use (15 years or more and at least 5 times/year) was 1.18. However, among postmenopausal women, the rates of uterine cancer were statistically significantly higher for those who commonly used hair relaxers even after adjustment for other potential risk factors. In postmenopausal women compared to never/light use, the Hazard Ratio for moderate use was 1.60, the Hazard Ratio for heavy use was 1.64, and the Hazard Ratio for 20 or more years of use regardless of frequency was 1.71. Postmenopausal women who reported using hair relaxers more than twice a year, or for more than five years had a greater than 50% increased risk of uterine cancer, compared to women who never or rarely used hair relaxers.
It was concluded from this large study that long-term use of chemical hair relaxers was associated with increased risk of uterine cancer among postmenopausal women, even after adjustment for other potential risk factors, but not among premenopausal women. These findings suggest that hair relaxer use may be a potentially modifiable risk factor for uterine cancer and the researchers hope these results will raise awareness of the potential toxic effects of these products and promote efforts to reduce exposure.
Hair relaxer use and risk of uterine cancer in the Black Women’s Health Study. Bertrand KA, Delp L, Coogan PF, et al. Environmental Research 2023. Volume 239, Part 1, 15 December 2023, 117228. https://doi.org/10.1016/j.envres.2023.117228