Dietary Supplement Use during Adjuvant Chemotherapy May Increase Risk for Breast Cancer Recurrence

SUMMARY: The Council for Responsible Nutrition reported that 77% of Americans consume dietary supplements. With the growing awareness regarding health, fitness and nutrition, the market size for dietary supplements is projected to hit a valuation of $349.4 billion by 2026.

Patients often use dietary supplements following a diagnosis of cancer, even though clinical recommendations discourage the use of antioxidant supplements during chemotherapy. One of the mechanisms of action of cytotoxic chemotherapeutic agents is through the generation of Reactive Oxygen Species (ROS). The use of dietary supplements during treatment, particularly antioxidants, could reduce the efficacy of cytotoxic agents. DELCaP study was conducted to address this concern.

DELCaP (Diet, Exercise, Lifestyle and Cancer Prognosis) trial is a prospective observational study, ancillary to an intergroup therapeutic clinical trial for high-risk breast cancer, conducted to evaluate associations between supplement use, particularly antioxidants during chemotherapy treatment, and breast cancer survival outcomes.

The Phase III SWOG S0221 trial evaluated the optimal dose and schedule of Anthracycline/Taxane adjuvant chemotherapy in women with high-risk early breast cancer. The current analysis involved a cohort of 1,134 of 2,014 patients enrolled in this study, who answered a baseline and follow-up questionnaires that included their use of dietary supplement at enrollment and during treatment. The authors then analyzed associations of dietary supplement use with clinical outcomes, after adjusting for clinical and lifestyle factors. Approximately 18% of patients used antioxidants such as Vitamins C, A, and E, Carotenoids or Coenzyme Q10 during treatment, whereas 44% of patients took multivitamins during chemotherapy.

It was noted from this analysis that the use of any antioxidant supplement (Vitamins A, C, and E, Carotenoids and Coenzyme Q10), both before and during adjuvant treatment was associated with an increased risk of recurrence versus no such use of supplements (HR=1.41; P=0.06). There was also a nonsignificant increased risk of overall mortality with the use of any antioxidant supplement (HR=1.40; P=0.14). There was a weaker relationship of outcomes with individual antioxidants and this may perhaps be due to the small numbers of patients. With regards to nonoxidants, Vitamin B12 use both before and during chemotherapy was significantly associated with poorer Disease Free Survival (HR=1.83; P<0.01) and Overall Survival (HR= 2.04; P<0.01). Use of iron during chemotherapy was also significantly associated with recurrence (HR=1.79; P<0.01), as was use both before and during treatment (HR=1.91; P=0.06). Results were similar for Overall Survival. Multivitamin use however was not associated with survival outcomes.

The researchers based on this analysis concluded that the use of antioxidant and nonantioxidant dietary supplements, but not multivitamins, before and during adjuvant chemotherapy may be associated with inferior treatment outcomes, in patients with early stage high risk breast cancer. They added that caution should be exercised by patients, when considering the use of supplements, other than a multivitamin, during chemotherapy, and patients should try to get their vitamins and minerals including antioxidants through food products..

Dietary Supplement Use During Chemotherapy and Survival Outcomes of Patients With Breast Cancer Enrolled in a Cooperative Group Clinical Trial (SWOG S0221). Ambrosone CB, Zirpoli GR, Hutson AD, et al. J Clin Oncol. 2019;38:804-814

FDA Approves ALUNBRIG® for First Line Treatment of ALK Positive Non Small Cell Lung Cancer

SUMMARY: The FDA on May 22, 2020 approved approved ALUNBRIG® (Brigatinib) for the first-line treatment of patients with ALK-positive metastatic Non Small Cell Lung Cancer (NSCLC), as detected by an FDA-approved test. Lung cancer is the second most common cancer in both men and women and accounts for about 14% of all new cancers and 27% of all cancer deaths. The American Cancer Society estimates that for 2020, about 228, 820 new cases of lung cancer will be diagnosed and 135,720 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 discovery of rearrangements of the Anaplastic Lymphoma Kinase (ALK) gene in some patients with advanced NSCLC and adenocarcinoma histology, led to the development of agents such as XALKORI® (Crizotinib), ZYKADIA® (Ceritinib), ALECENSA® (Alectinib) and ALUNBRIG® (Brigatinib), with promising results. It has become clear that appropriate, molecularly targeted therapy for tumors with a molecular abnormality, results in the best outcomes. According to the US Lung Cancer Mutation Consortium (LCMC), two thirds of patients with advanced adenocarcinoma of the lung, have a molecular driver abnormality. The most common oncogenic drivers in patients with advanced adenocarcinoma of the lung are, KRAS in 25%, EGFR in 21% and ALK in 8% as well as other mutations in BRAF, HER2, AKT1 and fusions involving RET and ROS oncogenes. These mutations are mutually exclusive, and the presence of two simultaneous mutations, are rare.

The new approval for ALUNBRIG® was based on results from the Phase III ALTA 1L (ALK in Lung Cancer Trial of BrigAtinib in 1st Line) trial, which is a global, ongoing, randomized, open-label, comparative, multicenter study, in which investigators compared the efficacy and safety of ALUNBRIG® with XALKORI® (Crizotinib) in 275 patients with Stage IIIB/IV ALK positive, locally advanced or metastatic NSCLC, who have not received prior treatment with an ALK inhibitor, but may have received 1 prior regimen of chemotherapy or no chemotherapy in the advanced setting. Patients were randomized 1:1 to receive either ALUNBRIG® 180 mg orally once daily (N=137), with a 7-day lead-in period at 90 mg, or XALKORI® 250 mg orally twice daily (N=138). Crossover from the XALKORI® arm to receive ALUNBRIG® was permitted at BICR (Blinded Independent Review Committee)-assessed Progression Free Survival (PFS). The median age was 59 years, and 55% of patients were female. Twenty-nine percent had brain metastases at baseline with comparable pre-enrollment central nervous system (CNS) radiotherapy rates among both cohorts. Overall, 27% of patients had prior chemotherapy in the locally advanced or metastatic setting. The Primary endpoint was BIRC assessed PFS and Secondary endpoints included Objective Response Rate (ORR), Intracranial ORR, Intracranial PFS, Overall Survival (OS), safety, and tolerability.

At a median follow up of 25 months, it was noted that ALUNBRIG® reduced the risk of disease progression or death by 51% compared with XALKORI® (HR=0.49; P=0.0007), with a median PFS of 24 months as assessed by a BIRC versus 11 months for XALKORI®. The confirmed ORR as assessed by BIRC was 74% with ALUNBRIG® and 62% for XALKORI®. The median duration of response (DOR) was not reached, and 13.8 months with ALUNBRIG® and XALKORI®, respectively.

After more than two years of follow-up, ALUNBRIG® demonstrated superiority over XALKORI®, with significant anti-tumor activity observed, especially in patients with baseline brain metastases. The confirmed intracranial ORR for patients with measurable brain metastases at baseline, treated with ALUNBRIG® was 78% versus 26% for patients treated with XALKORI®. The median intracranial Duration of Response in confirmed responders with measurable brain metastases at baseline was Not Reached with ALUNBRIG® and 9.2 months with XALKORI®, respectively. The median intracranial PFS was 24 months with ALUNBRIG®, compared with 5.6 months for XALKORI®. ALUNBRIG® reduced the risk of intracranial disease progression or death by 69% in patients who had brain metastases at baseline (HR=0.31).

Additionally, patients in the ALUNBRIG® group also experienced significant improvements in Health-Related Quality of Life, with delay in the median time to worsening in Global Health Score by 27 months versus 8 months with XALKORI®, as well as delay in the time to worsening and prolonged duration of improvement in fatigue, nausea and vomiting, appetite loss, and emotional and social functioning. Further, the duration of improvement in QoL with ALUNBRIG® was Not Reached versus 12 months with XALKORI®.

It was concluded that ALUNBRIG® demonstrated a statistically and clinically significant improvement in Progression Free Survival when compared to XALKORI® in ALK inhibitor-naïve, ALK positive NSCLC, with superior efficacy especially among those with brain metastases at baseline.

Brigatinib vs crizotinib in patients with ALK inhibitor-naive advanced ALK+ NSCLC: Updated results from the phase III ALTA-1L trial. Camidge R, Kim HR, Ahn M, et al. Presented at the 2019 ESMO Asia Congress, November 23, 2019.

FDA Approves Chemotherapy-Free First Line Immunotherapy Combination in Advanced NSCLC

SUMMARY: The FDA on May 15, 2020, approved OPDIVO® (Nivolumab) in combination with YERVOY® (Ipilimumab), as first-line treatment for patients with metastatic Non-Small Cell Lung Cancer (NSCLC), whose tumors express PD-L1(1% or more), as determined by an FDA-approved test, with no Epidermal Growth Factor Receptor (EGFR) or Anaplastic Lymphoma Kinase (ALK) genomic tumor aberrations. Lung cancer is the second most common cancer in both men and women and accounts for about 14% of all new cancers and 27% of all cancer deaths. The American Cancer Society estimates that for 2020, about 228, 820 new cases of lung cancer will be diagnosed and 135,720 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.

Immune checkpoints are cell surface inhibitory proteins/receptors that are expressed on activated T cells. They harness the immune system and prevent uncontrolled immune reactions by switching off the immune system T cells. Immune checkpoint proteins/receptors include CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4, also known as CD152) and PD-1(Programmed cell Death 1). Checkpoint inhibitors unleash the T cells resulting in T cell proliferation, activation, and a therapeutic response. OPDIVO® (Nivolumab) is a fully human, immunoglobulin G4 monoclonal antibody that binds to the PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, thereby undoing PD-1 pathway-mediated inhibition of the immune response and unleashing the T cells. OPDIVO® is presently approved for treatment of patients with metastatic NSCLC and progression on or after Platinum-based chemotherapy. YERVOY® is a fully human immunoglobulin G1 monoclonal antibody that blocks Immune checkpoint protein/receptor CTLA-4.

The present FDA approval was based on CheckMate-227, which is an open-label, multi-part, global, Phase III trial in which OPDIVO® based regimens were compared with Platinum-doublet chemotherapy in patients with first line advanced NSCLC, across non-squamous and squamous tumor histologies. In Part 1 of this trial, there were 2 cohortsPart 1a in which OPDIVO® plus low dose YERVOY® (N=396) or OPDIVO® monotherapy (N=396) was compared with chemotherapy alone (N=397), in patients whose tumors expressed PD-L1 expression of 1% or more and Part 1b in which OPDIVO® plus low dose YERVOY® (N=187) or OPDIVO® plus chemotherapy (N=177) was compared with chemotherapy alone (N=186), in patients whose tumors did not express PD-L1 (less than 1%). (In Part 2 of this trial, OPDIVO® plus chemotherapy was compared with chemotherapy alone, regardless of PD-L1 expression. Part 2 did not meet its Primary endpoint for Overall Survival for OPDIVO® plus chemotherapy versus chemotherapy alone, in patients with non-squamous NSCLC, and is published elsewhere). It should be noted that when this trial was launched, chemotherapy along with immunotherapy or immunotherapy alone was not approved for the front-line treatment of NSCLC. Therefore, dual immunotherapy combination was not compared with current standards of care such as chemotherapy plus immunotherapy.

OPDIVO® was administered at 3 mg/kg every 2 weeks, and in the combination arm, YERVOY® was administered at 1 mg/kg every 6 weeks. When administered with chemotherapy, OPDIVO® was administered at 360 mg every 3 weeks. Patients were stratified by histology, and treatment was administered until disease progression, unacceptable toxicity, or administered for 2 years for immunotherapy. There were two Co-primary endpoints in Part 1 for OPDIVO® plus YERVOY® versus chemotherapy: Overall survival (OS) in patients whose tumors express PD-L1 (assessed in patients enrolled in Part 1a) and Progression Free Survival (PFS) in patients with TMB of 10 mut/Mb or more, across the PD-L1 spectrum (assessed in patients enrolled across Parts 1a and 1b). The minimum follow up for the Primary endpoint was 29 months. Both Part 1a and Part 1b groups met their Primary endpoints.

In the Part 1a cohort with PD-L1 expression of 1% or more, the Overall Survival was significantly longer with OPDIVO® plus YERVOY®, compared to chemotherapy. The median Overall Survival was 17.1 months in the Immunotherapy combination group compared to 14.9 months in the chemotherapy group (HR=0.79; P=0.007), with a 2-year OS rates of 40.0% and 32.8%, respectively. Progression Free Survival, Objective Response Rates and Duration of Response were all greater with OPDIVO® plus YERVOY® combination, compared to chemotherapy. The median Progression Free Survival (PFS) was 5.1 months in the OPDIVO® plus YERVOY® group and 5.6 months in the platinum-doublet chemotherapy group (HR=0.82). Confirmed Overall Response Rate (ORR) was 36% and 30% respectively. Median Duration of Response was 23.2 months in the OPDIVO® plus YERVOY® group and 6.2 months in the platinum-doublet chemotherapy group. In the Part 1b cohort with PD-L1 expression of less than 1%, Overall Survival benefit was again observed with the OPDIVO® plus YERVOY® combination, compared with chemotherapy, with a median duration of 17.2 months with OPDIVO® plus YERVOY® and 12.2 months with chemotherapy. Among all the patients in the trial, the median duration of OS was 17.1 months with OPDIVO® plus YERVOY® and 13.9 months with chemotherapy. Grade 3 and 4 treatment-related Adverse Events across all patients was 33% in those treated with OPDIVO® plus YERVOY® combination and 36% with chemotherapy.

It was concluded that first-line treatment of patients with advanced NSCLC, with a combination of two immunotherapy drugs, improves Overall Survival, compared to chemotherapy, independent of the PD-L1 expression level, and offers a chemotherapy-free first line treatment option for a subset of NSCLC patients, leaving chemotherapy for later lines of therapy.

Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. N Engl J Med. 2019;381:2020-2031.

FDA Approves RUBRACA® for BRCA-Mutated Metastatic Castrate Resistant Prostate Cancer

SUMMARY: The FDA on May 15, 2020, granted accelerated approval to RUBRACA® (Rucaparib) for patients with deleterious BRCA mutation (germline and/or somatic)-associated metastatic Castration-Resistant Prostate Cancer (mCRPC), who have been treated with Androgen Receptor-directed therapy and a taxane-based chemotherapy. Prostate cancer is the most common cancer in American men with the exclusion of skin cancer, and 1 in 9 men will be diagnosed with prostate cancer during their lifetime. It is estimated that in the United States, about 191,930 new cases of prostate cancer will be diagnosed in 2020 and 33,330 men will die of the disease.

The development and progression of Prostate cancer is driven by androgens. Androgen Deprivation Therapy (ADT) or testosterone suppression has therefore been the cornerstone of treatment of advanced Prostate cancer and is the first treatment intervention. Androgen Deprivation Therapies have included bilateral orchiectomy or Gonadotropin Releasing Hormone (GnRH) analogues, with or without first generation Androgen Receptor (AR) inhibitors such as CASODEX® (Bicalutamide), NILANDRON® (Nilutamide) and EULEXIN® (Flutamide) or with second-generation, anti-androgen agents, which include, ZYTIGA® (Abiraterone), XTANDI® (Enzalutamide) and ERLEADA® (Apalutamide). ZYTIGA® inhibits CYP17A1 enzyme and depletes adrenal and intratumoral androgens, thereby impairing AR signaling. XTANDI® and ERLEADA® compete with Testosterone and Dihydrotestosterone and avidly bind to the Androgen Receptor, thereby inhibiting AR signaling, and in addition inhibit translocation of the AR into the nucleus and thus inhibits the transcriptional activities of the AR. Approximately 10-20% of patients with advanced Prostate cancer will progress to Castration Resistant Prostate Cancer (CRPC) within five years during ADT, and over 80% of these patients will have metastatic disease at the time of CRPC diagnosis. The estimated mean survival of patients with CRPC is 9-36 months, and there is therefore an unmet need for new effective therapies.

DNA damage is a common occurrence in daily life by UV light, ionizing radiation, replication errors, chemical agents, etc. This can result in single and double strand breaks in the DNA structure which must be repaired for cell survival. The two vital pathways for DNA repair in a normal cell are BRCA1/BRCA2 and PARP. The PARP (Poly ADP Ribose Polymerase), family of enzymes include, PARP1, PARP2 and PARP3. BRCA1 and BRCA2 genes recognize and repair double strand DNA breaks via Homologous Recombination (HR) pathway. Homologous Recombination is a type of genetic recombination, and is a DNA repair pathway utilized by cells to accurately repair DNA double-stranded breaks during the S and G2 phases of the cell cycle, and thereby maintain genomic integrity. Homologous Recombination Deficiency (HRD) is noted following mutation of genes involved in HR repair pathway. At least 15 genes are involved in the Homologous Recombination Repair (HRR) pathway including BRCA1, BRCA2 and ATM genes. The BRCA1 gene is located on the long (q) arm of chromosome 17 whereas BRCA2 is located on the long arm of chromosome 13. BRCA1 and BRCA2 are tumor suppressor genes and functional BRCA proteins therefore repair damaged DNA, and play an important role in maintaining cellular genetic integrity. They regulate cell growth and prevent abnormal cell division and development of malignancy.

Recently published data has shown that deleterious germline and/or somatic mutations in BRCA1, BRCA2, ATM, or other Homologous Recombination DNA-repair genes, are present in about 25% of patients with advanced prostate cancer, including mCRPC. Mutations in BRCA1 and BRCA2 also account for about 20-25% of hereditary breast cancers, about 5-10% of all breast cancers, and 15% of ovarian cancers. BRCA mutations can either be inherited (Germline) and present in all individual cells or can be acquired and occur exclusively in the tumor cells (Somatic). Somatic mutations account for a significant portion of overall BRCA1 and BRCA2 aberrations. Loss of BRCA function due to frequent somatic aberrations likely deregulates HR pathway, and other pathways then come in to play, which are less precise and error prone, resulting in the accumulation of additional mutations and chromosomal instability in the cell, with subsequent malignant transformation. HRD therefore indicates an important loss of DNA repair function. PARP is a related enzymatic pathway that repairs single strand breaks in DNA. In a BRCA mutant, the cancer cell relies solely on PARP pathway for DNA repair. In the presence of a PARP inhibitor, there is synthetic lethality because of the loss of both repair pathway genes, leading to cell death. Thus PARP inhibitors are only harmful to cancer cells.MOA-of-RUBRACA

RUBRACA® is an oral, small molecule inhibitor of Poly-Adenosine diphosphate [ADP] Ribose Polymerase (PARP), presently approved by the FDA for ovarian, fallopian tube or primary peritoneal cancers. This recent FDA approval for prostate cancer patients was based on TRITON2, which is an ongoing international, multicenter, open-label, single arm, Phase II trial, in patients with BRCA-mutated mCRPC, who had been treated with Androgen Receptor-directed therapy and taxane-based chemotherapy. In this study, 115 mCRPC patients with either germline or somatic BRCA mutations were enrolled, of whom 62 patients had measurable disease at baseline. Patients received RUBRACA® 600 mg orally twice daily and concomitant GnRH analog or had prior bilateral orchiectomy. Treatment was continued until disease progression or unacceptable toxicity. The median age was 73 years, majority of patients had an ECOG performance status of 0 or 1, 18% of patients had lung metastases, 21% had liver metastases, 24% had metastases to lymph nodes alone and 40% had 10 or more bone lesions at baseline. The major efficacy outcomes of the study were Objective Response Rate (ORR) and Duration of Response (DOR) in the 62 patients with measurable disease. The median duration of follow up was 13.1 months

The confirmed ORR was 44% and the median DOR was not evaluable. Fifty six percent (56%) of patients with confirmed Objective Responses had a DOR of 6 months or more.

It was concluded that RUBRACA® demonstrates promising efficacy in patients with mCRPC with deleterious BRCA mutations. TRITON3 study is evaluating RUBRACA® versus physician’s choice of second-line AR-directed therapy or Docetaxel, in chemotherapy-naïve patients with mCRPC and alterations in BRCA1/2, who progressed on one prior AR-directed therapy.

ESMO 2019: Preliminary Results from the TRITON2 Study of Rucaparib in Patients with DNA Damage Repair-deficient mCRPC: Updated Analyses. Abida W, Campbell D, Patnaik A, et al. 2019 ESMO Annual Meeting, #ESMO19, 27 Sept – 1 Oct 2019 in Barcelona, Spain.

FDA Approves TABRECTA® for Metastatic Non-Small Cell Lung Cancer

SUMMARY: The FDA on May 6, 2020, granted accelerated approval to TABRECTA® (Capmatinib) for adult patients with metastatic Non-Small Cell Lung Cancer (NSCLC), whose tumors have a mutation that leads to Mesenchymal-Epithelial Transition (MET) exon 14 skipping, as detected by an FDA-approved test. The FDA also approved the FoundationOne CDx assay (Foundation Medicine, Inc.) as a companion diagnostic for TABRECTA®.

MET is a widely expressed Receptor Tyrosine Kinase and plays a pivotal role in cell growth, proliferation and survival. The MET gene encodes for a protein known as the Hepatocyte Growth Factor (HGF) Receptor. Upon binding by Hepatocyte Growth Factor (HGF), the HGF Receptor is activated, with resulting activation of the downstream RAS/RAF/MEK/ERK and PI3K/AKT/mTOR signaling pathways, thereby serving different important biological functions. Alterations in the MET gene leading to abnormal MET signaling, has been identified in different types of cancers including thyroid, lung, breast, liver, colon, kidney, ovary and gastric carcinoma.MET-Signaling-Pathway

Two key MET alterations include MET exon 14 skipping mutations and MET amplification. MET exon 14 skipping mutations occur in approximately 5% of NSCLC patients with enrichment in sarcomatoid lung cancers (22%). MET exon 14 skipping mutation is a recognized oncogenic driver and is a molecular genetic abnormality indicating the presence of a splice site mutation that results in a loss of transcription of exon 14 of the MET gene. Most exon 14 mutations occur in never-smokers and is seen in both squamous and adenocarcinoma histology. Patients whose cancers have MET exon 14 skipping generally have very high response rates to MET inhibitors and molecular testing for MET exon 14 skipping should therefore be performed on all lung cancers, because this is a targetable alteration. MET amplification has been more commonly seen in smokers, and responses in patients with MET-amplified tumors might be more variable and dependent on level of amplification, with higher responses noted in tumors with more than 5-6 fold amplification. Tumors with MET exon 14 skipping mutations usually do not harbor activating mutations in EGFR, KRAS, or BRAF or concurrent ALK, ROS1 or RET translocations. However, it appears that cMET exon 14 skipping is not mutually exclusive with cMET amplification.

TABRECTA® (Capmatinib) is a highly potent and selective, reversible inhibitor of MET tyrosine kinase. The present FDA approval was based on the primary findings from the Phase II GEOMETRY mono-1 trial, which is a non-randomized, open-label, multi-cohort, Phase II study, conducted to evaluate the efficacy and safety of single-agent TABRECTA® in adult patients with EGFR wild-type, ALK-negative, metastatic NSCLC, whose tumors have a mutation that leads to MET exon 14 skipping (METex14), as detected by an RNA-based RT-PCR. This study enrolled 97 patients with metastatic NSCLC and confirmed MET exon 14 skipping mutations, 69 of whom were previously treated and, 28 of whom, were treatment naive. The patients received TABRECTA® at 400 mg orally twice daily until disease progression or unacceptable toxicity. The median patient age was 71 years and all NSCLC histologies including sarcomatoid/carcinosarcoma were included. Majority of the patients (75%) were white and 24% were Asian. Previous treatments included immunotherapy (28%) and chemotherapy (94%), and 23% of patients received 2 prior lines of therapy. The main efficacy outcome was Overall Response Rate (ORR) and additional efficacy outcomes included Duration of Response, Time to Response, Disease Control Rate, Progression Free Survival (PFS) and Safety. Thirteen patients (N=13) in this study had brain metastases at baseline.

Among the treatment-naïve patients group, the ORR was 68% with a median Duration of Response of 12.6 months and the percentage of patients with responses for 12 months or longer was 47%. The Disease Control Rate (Complete Response plus Partial Response plus Stable Disease) was 96.4%.

Among the previously treated patients, the ORR was 41%, with a median Duration of Response of 9.7 months and the percentage of patients with responses for 12 months or longer was 32%. The Disease Control Rate was 78.3%. Among those with brain metastases at baseline, 54% had an intracranial response with TABRECTA® with 31% showing complete resolution, 23% showing partial resolution, and the intracranial Disease Control Rate was 92%. The most common adverse events (occurring in at least 20% of patients) were peripheral edema, nausea, fatigue, vomiting, dyspnea, and decreased appetite. TABRECTA® can also cause Interstitial Lung Disease, hepatotoxicity and photosensitivity.

It was concluded that TABRECTA® is a new treatment option for patients with MET exon 14 skipping- mutated advanced NSCLC, regardless of the line of therapy, with deep and durable responses, manageable toxicity profile, and is the first and only FDA approved treatment for this patient group.
Capmatinib (INC280) in METex14-mutated advanced non-small cell lung cancer (NSCLC): Efficacy data from the phase II GEOMETRY mono-1 study. Wolf J, Seto T, Han J, et al. J Clin Oncol. 2019;37(suppl; abstr 9004).

Proton Based Chemoradiotherapy Significantly Decreases Toxicities without Compromising Efficacy

SUMMARY: Radiation Therapy involves the use of X-Rays, Gamma rays and charged particles for cancer treatment. External Beam Radiation Therapy (EBRT) is most often delivered using a linear accelerator in the form of Photon beams (either X-rays or Gamma rays). Photons have no mass and are packets of energy of an electromagnetic wave. Electrons and Protons are charged particles and Electrons are considered light particles whereas Protons are considered heavy particles. Electron beams are used to irradiate skin and superficial tumors, as they are unable to penetrate deep into the tissues. The different types of External Beam Radiation Treatments include 3-Dimensional Conformal Radiation Therapy (3D-CRT) meant to deliver radiation to very precisely shaped target areas, IMRT or Intensity Modulated Radiation Therapy which allows different areas of a tumor or nearby tissues to receive different doses of radiation, Image Guided Radiation Therapy (IGRT) which allows reduction in the planned volume of tissue to be treated, as changes in a tumor size are noted during treatment, Stereotactic RadioSurgery (SRS) which can deliver one or more high doses of radiation to a small tumor and Stereotactic Body Radiation Therapy (SBRT) or CYBERKNIFE® which is similar to SRS but also takes the normal motion of the body into account while treating malignancies involving the lung and liver.

Proton beams unlike Photons, enter the skin and travel through the tissues and deposit much of their energy at the end of their path (known as the Bragg peak), and deposit less energy along the way. This is unlike Photons which deposit energy all along the path through the tissues and the deposited dose decreases with increasing depth. As a result, with Proton beam therapy, normal tissues are exposed to less radiation compared with Photons. Despite this advantage, tissue heterogeneity such as organ motion, tumor volume changes during treatment can have a significant negative impact on target coverage for Proton beam therapy and can result in damage to the surrounding tissues and potential complications. It is well established that there is significant benefit for Proton beam therapy in certain pediatric malignancies.Types-of-Radiation-Therapy

Curative treatment with concurrent chemoradiotherapy is the standard of care for many nonmetastatic, locally advanced cancers. This treatment modality however is associated with substantial morbidity. Proton therapy as component of concurrent chemoradiotherapy might be able to reduce treatment related toxicity and achieve comparable cancer control outcomes, compared with conventional Photon radiotherapy, by reducing the radiation dose to normal tissues. There are however limited data comparing results of Proton chemoradiotherapy with Photon chemoradiotherapy, and Proton therapy remains unproven in this treatment setting. The objective of this study was to assess whether Proton therapy in the setting of concurrent chemoradiotherapy is associated with fewer hospitalizations or other adverse events and similar Disease-free and Overall Survival, compared with concurrent Photon chemoradiotherapy.

In this large single-institution, nonrandomized, comparative effectiveness, retrospective analysis, 1483 adult patients with nonmetastatic, locally advanced cancer, treated with concurrent chemoradiotherapy with curative intent were included. Three hundred ninety-one patients (N=391) received Proton therapy and 1092 patients received Photon therapy. Common tumor sites included head and neck, lung, brain, esophagus/gastric, rectum, and pancreas. The median patient age was 62 years, but patients treated with Protons were significantly older with a median age of 66 years versus 61 years, had less favorable Charlson-Deyo comorbidity scores and had lower integral radiation dose to tissues outside the target. Ninety three percent (93%) of patients in the Photon therapy group were treated with Intensity-Modulated Radiotherapy (IMRT). Baseline ECOG Performance Status was similar between the two treatment cohorts. The Primary end point was 90-day adverse events associated with unplanned hospitalizations (CTCAE version 4 – Grade 3 or more). Secondary end points included ECOG performance status decline during treatment, 90-day adverse events of at least Grade 2 that limit instrumental activities of daily living, and Disease-Free and Overall Survival. The data on adverse events and survival were gathered prospectively.

It was noted that Proton chemoradiotherapy was associated with a significantly lower relative risk of 90-day adverse events of at least Grade 3 (P=0.002), significantly lower relative risk of 90-day adverse events of at least Grade 2 (P=0.006), and decline in Performance Status during treatment (P<0.001). Proton chemoradiotherapy was associated with a two-thirds reduction in adverse events associated with unplanned hospitalizations. At a median follow up of 3.7 years for the Proton cohort and 4.2 years for the Photon cohort, there was no difference in Disease-Free or Overall Survival.

It was concluded from this analysis that in adults with locally advanced cancer, Proton chemoradiotherapy was associated with significantly reduced acute adverse events that caused unplanned hospitalizations, with similar Disease-Free and Overall Survival, compared to Photon therapy.
Comparative Effectiveness of Proton vs Photon Therapy as Part of Concurrent Chemoradiotherapy for Locally Advanced Cancer. Baumann BC, Mitra N, Harton JG, et al. Jama Oncol. 2020;6:237-246.

FDA Approves ZEJULA® for Newly Diagnosed Patients with Advanced Ovarian Cancer

SUMMARY: The FDA on April 29, 2020 approved ZEJULA® (Niraparib) for the maintenance treatment of adult patients with advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer, who are in a complete or partial response to first-line platinum-based chemotherapy. It is estimated that in the United States, approximately 21,750 women will be diagnosed with ovarian cancer in 2020 and 13,940 women will die of the disease. Ovarian cancer ranks fifth in cancer deaths among women, and accounts for more deaths than any other cancer of the female reproductive system. Approximately 75% of the ovarian cancer patients are diagnosed with advanced disease. Patients with newly diagnosed advanced ovarian cancer are often treated with platinum based chemotherapy following primary surgical cytoreduction. Approximately 70% of these patients will relapse within the subsequent 3 years and are incurable, with a 5 year Overall Survival rate of about 20-30%.MOA-of-PARP-Inhibitors

DNA damage is a common occurrence in daily life by UV light, ionizing radiation, replication errors, chemical agents, etc. This can result in single and double strand breaks in the DNA structure which must be repaired for cell survival. The two vital pathways for DNA repair in a normal cell are BRCA1/BRCA2 and PARP. The PARP (Poly ADP Ribose Polymerase) family of enzymes, include PARP1 and PARP2. In the context of DNA repair, BRCA1 and BRCA2 genes recognize and repair double strand DNA breaks via Homologous Recombination (HR) pathway. Homologous Recombination is a type of genetic recombination, and is a DNA repair pathway utilized by cells to accurately repair DNA double-stranded breaks during the S and G2 phases of the cell cycle, and thereby maintain genomic integrity. Homologous Recombination Deficiency (HRD) is noted following mutation of genes involved in HR repair pathway. At least 15 genes are involved in the Homologous Recombination Repair (HRR) pathway including BRCA1 and BRCA2 genes. The BRCA1 gene is located on the long (q) arm of chromosome 17 whereas BRCA2 is located on the long arm of chromosome 13. BRCA1 and BRCA2 are tumor suppressor genes and functional BRCA proteins repair damaged DNA, and play an important role in maintaining cellular genetic integrity. They regulate cell growth and prevent abnormal cell division and development of malignancy. Mutations in BRCA1 and BRCA2 account for about 20-25% of hereditary breast cancers and about 5-10% of all breast cancers. They also account for 15% of ovarian cancers, in addition to other cancers such as Colon and Prostate. BRCA mutations can either be inherited (Germline) and present in all individual cells or can be acquired and occur exclusively in the tumor cells (Somatic). Somatic mutations account for a significant portion of overall BRCA1 and BRCA2 aberrations. Loss of BRCA function due to frequent somatic aberrations likely deregulates HR pathway, and other pathways then come in to play, which are less precise and error prone, resulting in the accumulation of additional mutations and chromosomal instability in the cell, with subsequent malignant transformation. HRD therefore indicates an important loss of DNA repair function. Hereditary Epithelial Ovarian Cancer was thought to be caused almost exclusively by mutations in BRCA1 and BRCA2. It however is now well known that about 50% of the high grade serous ovarian cancers have aberrations in HR repair pathway. Deregulated HR pathway increases sensitivity to platinum drugs. Majority of the women with germline BRCA mutations (gBRCA) are positive for HR deficiency.

PARP is a related enzymatic pathway that repairs single strand breaks in DNA. In a BRCA mutant, the cancer cell relies solely on PARP pathway for DNA repair. In the presence of a PARP inhibitor, there is synthetic lethality because loss of both genes, leading to cell death. Thus PARP inhibitors are only harmful to cancer cells. ZEJULA® is a highly selective PARP 1/2 inhibitor, that causes cumulative DNA damage and cell death by inhibiting PARP. Previously published phase III study among patients with platinum-sensitive, recurrent ovarian cancer (NEJM 2016;375:2154-2164) concluded that Niraparib significantly prolonged Progression Free Survival (PFS) compared to placebo, and this benefit was achieved regardless of the presence or absence of germline BRCA mutations or HRD status.

PRIMA trial is a randomized, double-blind, placebo-controlled, international Phase III trial conducted to test the efficacy and safety of ZEJULA® maintenance therapy after a response to platinum-based chemotherapy, in patients with newly diagnosed advanced ovarian cancer at high risk for relapse. It should be noted that at the time PRIMA trial was designed, AVASTIN® (Bevacizumab) was not approved for first-line treatment in all participating countries. A total of 733 patients with newly diagnosed, high risk, advanced ovarian cancer were randomly assigned in a 2:1 ratio to receive ZEJULA® (N=487) or placebo (N=246) once daily in 28-day cycles for 36 months or until disease progression, after a response to platinum-based chemotherapy regimen. Patients received a dose of 200-300mg once daily, based on body weight and platelet count. Enrolled patients were at high risk for progressive disease with 23.1% having Stage III ovarian cancer with residual disease after primary debulking surgery, 66.7% had received neoadjuvant chemotherapy, 35% had Stage IV ovarian cancer, and 30.5% had a Partial Response to first-line platinum-based chemotherapy. Tumor samples were tested for HRD status and HRD was defined by either presence of tumor BRCA mutation or Genomic Instability Score (GIS) of 42 or more. Of the randomized patients, 50.9% had tumors with HRD, 30.4% had BRCA mutations and 20.5% were BRCA wild type. The treatment groups were well balanced. The Primary endpoint was Progression Free Survival (PFS) in patients who had tumors with HRD, and then in the overall population, as determined on hierarchical testing. Secondary end points included Overall Survival, time until the first subsequent therapy, PFS 2, defined as time from randomization to progression while the patient was receiving a subsequent anticancer therapy and Patient-Reported Outcomes. The median duration of follow-up at the time of the data cutoff was 13.8 months.

There was a statistically significant improvement in PFS for patients randomized to ZEJULA® compared with placebo in the HRD group, as well as the overall population. The median PFS in the HRD group was 21.9 months for patients receiving ZEJULA® compared with 10.4 months for those receiving placebo (HR=0.43; P<0.001). The median PFS in the overall population was 13.8 months for patients receiving ZEJULA® compared with 8.2 months for those receiving placebo (HR=0.62; P<0.001). At the 24-month interim analysis, the rate of Overall Survival was 84% in the ZEJULA® group and 77% in the placebo group (HR=0.70). The most common adverse reactions in patients receiving ZEJULA® were cytopenias, fatigue, AST/ALT elevation, hypertension, low grade nausea and decreased appetite.

It was concluded that among patients with newly diagnosed advanced ovarian cancer who had responded to platinum-based chemotherapy, ZEJULA® significantly prolonged Progression Free Survival, compared to those who received placebo, regardless of the presence or absence of Homologous Recombination Deficiency.
Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. González-Martín A, Pothuri B, Vergote I, et al. for the PRIMA/ENGOT-OV26/GOG-3012 Investigators. N Engl J Med 2019; 381:2391-2402

FDA Approves Subcutaneous DARZALEX® in Multiple Myeloma

SUMMARY: The FDA on May 1, 2020 approved DARZALEX® (Daratumumab) and Hyaluronidase-fihj (DARZALEX FASPRO®), for adult patients with newly diagnosed or Relapsed/Refractory multiple myeloma. This new product allows for subcutaneous dosing of DARZALEX®.

DARZALEX FASPRO® is now approved for these previously approved indications for IV DARZALEX®
1) In combination with VELCADE® (Bortezomib), Melphalan and Prednisone in newly diagnosed patients who are ineligible for Autologous Stem Cell Transplant (ASCT)
2) In combination with REVLIMID® (Lenalidomide) and Dexamethasone in newly diagnosed patients, who are ineligible for ASCT and in patients with Relapsed or Refractory multiple myeloma who have received at least one prior therapy
3) In combination with VELCADE® and Dexamethasone in patients who have received at least one prior therapy
4) As monotherapy, in patients who have received at least three prior lines of therapy including a Proteasome Inhibitor (PI) and an Immunomodulatory agent or who are double-refractory to a PI and an immunomodulatory agent.

DARZALEX® is a human IgG1 antibody that targets CD38, a transmembrane glycoprotein abundantly expressed on malignant plasma cells and with low levels of expression on normal lymphoid and myeloid cells. DARZALEX® exerts its cytotoxic effect on myeloma cells by multiple mechanisms, including Antibody Dependent Cellular Cytotoxicity (ADCC), Complement Dependent Cytotoxicity (CDC) and direct Apoptosis. Additionally, DARZALEX® may play a role in immunomodulation, by depleting CD38-positive regulator immune suppressor cells, and thereby expanding T cells, in patients responding to therapy.Mechanism-of-Action-of-Daratumumab

This FDA approval is based on COLUMBA Trial, which is a randomized, open-label, multicenter Phase III study, which included 522 patients with multiple myeloma, who had received at least three prior lines of therapy including a Proteasome Inhibitor (PI) and an immunomodulatory drug (IMiD), or whose disease was refractory to both a PI and an IMiD. Patients were randomly assigned to receive a fixed dose of subcutaneously (SC) administered formulation of DARZALEX® 1800 mg weekly for cycles 1-2, every two weeks for cycles 3-6 and every four weeks for cycle 7 and thereafter (N=263), with the subcutaneous preparation given over 3-5 minutes at alternating left and right abdominal sites. In the intravenous group, patients received DARZALEX® 16 mg/kg IV weekly for cycles 1-2, every two weeks for cycles 3-6 and every four weeks for cycle 7 and thereafter (N=259). Each cycle was 28 days. Treatment in both patient groups was continued until disease progression or unacceptable toxicity. The median age was 67 years and the median number of prior therapies was four in each treatment group. Patient characteristics were similar between the two arms except that more patients in the subcutaneous arm had high-risk cytogenetics (26%) compared with the intravenous group (17%). The median duration of treatment was approximately 5 months, with a median of 6 completed cycles of treatment. The median duration of infusion was consistently 5 minutes at each visit in the subcutaneous group. However, in the IV arm, the first infusion lasted 7 hours, the second infusion was 4.3 hours, and subsequent infusions lasted a median of 3.4 hours. The study co-Primary endpoints were Overall Response Rate (ORR) and pharmacokinetic endpoint of the maximum C-trough on cycle 3, day 1 pre-dose.

At a median follow up of 7.5 months, the ORR was 41% for the subcutaneous administered formulation of DARZALEX® compared to 37% for IV DARZALEX® (P<0.0001). The ORR was similar across all clinically relevant subgroups, including body weight. The ratio of geometric means of C-trough for the SC administered formulation of DARZALEX® over IV DARZALEX® was 108%. The Progression Free Survival was comparable between the SC administered formulation of DARZALEX and the current IV formulation of DARZALEX (HR=0.99; P<0.9258). A lower rate of infusion-related reactions was observed in the group that received the SC DARZALEX® compared to IV DARZALEX® (13% vs. 35%, respectively).

It was concluded that the subcutaneous formulation of DARZALEX® resulted in non-inferior pharmacokinetics and efficacy compared to the current IV formulation, and also importantly offers the potential for a fixed-dose administration, shorter administration times and a lower rate of infusion-related reactions with improved safety profile, in patients with Relapsed or Refractory multiple myeloma.

Subcutaneous versus intravenous daratumumab in patients with relapsed or refractory multiple myeloma (COLUMBA): a multicentre, open-label, non-inferiority, randomised, phase 3 trial. Mateos M-V, Nahi H, Legiec W, et al. The Lancet Haematology. Published: March 23, 2020. DOI: https://doi.org/10.1016/S2352-3026(20)30070-3.

FDA Approves TRODELVY® for Advanced Triple Negative Breast Cancer

SUMMARY: The FDA on April 22, 2020, granted accelerated approval to TRODELVY® (Sacituzumab govitecan-hziy), for adult patients with metastatic Triple-Negative Breast Cancer (TNBC), who received at least two prior therapies for metastatic disease. Breast cancer is the most common cancer among women in the US and about 1 in 8 women (13%) will develop invasive breast cancer during their lifetime. Approximately 276,480 new cases of invasive female breast cancer will be diagnosed in 2020 and about 42,170 women will die of the disease.

Triple Negative Breast Cancer (TNBC) is a heterogeneous, molecularly diverse group of breast cancers and are ER (Estrogen Receptor), PR (Progesterone Receptor) and HER2 (Human Epidermal Growth Factor Receptor-2) negative. TNBC accounts for 15-20% of invasive breast cancers, with a higher incidence noted in young patients and African American females. It is usually aggressive, and tumors tend to be high grade, and patients with TNBC are at a higher risk of both local and distant recurrence and often develop visceral metastases. Those with metastatic disease have one of the worst prognoses of all cancers with a median Overall Survival of 13 months. The majority of patients with TNBC who develop metastatic disease do so within the first 3 years after diagnosis, whereas those without recurrence during this period of time have survival rates similar to those with ER-positive breast cancers. The lack of known recurrent oncogenic drivers in patients with metastatic TNBC, presents a major therapeutic challenge. Overall survival among patients with pretreated metastatic TNBC has not changed over the past 2 decades and standard chemotherapy is associated with low response rates of 10-15% and a Progression Free Survival of only 2-3 months.

TRODELVY® is an Antibody-Drug Conjugate (ADC) in which SN-38, an active metabolite of Irinotecan, a Topoisomerase I inhibitor, is coupled to the humanized Anti-Trophoblast cell-surface antigen 2 (Trop-2) monoclonal antibody (hRS7 IgG1κ), through the cleavable CL2A linker. SN-38 cannot be given directly to patients because of its toxicity and poor solubility. Trop-2, a transmembrane calcium signal transducer, stimulates cancer-cell growth, and this cell surface receptor is overexpressed in several epithelial cancers including cancers of the breast, colon and lung, and has limited expression in normal human tissues. Trop-2 is expressed in more than 85% of breast tumors including Triple Negative Breast Cancer. Upon binding to Trop-2, the anti-TROP-2 monoclonal antibody is internalized and delivers SN-38 directly into the tumor cell, making it a suitable transporter for the delivery of cytotoxic drugs. Further, the cleavable linker enables SN-38 to be released both intracellularly into the tumor cells as well as the tumor microenvironment, thereby allowing for the delivery of therapeutic concentrations of the active drug in bystander cells to which the conjugate has not bound. Thus, TRODELVY®-bound tumor cells are killed by intracellular uptake of SN-38, whereas the adjacent tumor cells are killed by the extracellular release of SN-38.

IMMU-132-01 is a Phase I/II, basket design, open-label, single-group, multicenter trial involving patients with various types of advanced epithelial cancers, who have received at least one previous therapy for metastatic disease. (One example of a basket design is a single drug evaluated in multiple baskets, with each basket representing a different malignancy or tumor site with the same target). A total of 108 patients with metastatic Triple-Negative Breast Cancer (TNBC) were enrolled between June 2013 and February 2017. Patients received TRODELVY® 10 mg/kg IV on days 1 and 8 every 21 days. Tumor imaging was obtained every 8 weeks, and patients were treated until disease progression or intolerance to therapy. The median patient age was 55 years. Enrolled patients had a median of 3 prior anticancer regimens and 98% had received taxanes and 86% had received anthracyclines. The Primary efficacy end point was the Objective Response Rate (ORR). Other efficacy end points included Time to Response and Duration of Response in patients who had a response, the Clinical Benefit Rate (defined as a Complete or Partial Response or stable disease for at least 6 months), Progression Free and Overall Survival. The median duration of follow up for this basket of 108 patients with metastatic TNBC was 9.7 months.

The Objective Response Rate was 33.3% including a Complete Response Rate of 2.8%. The median Time to Response was 2.0 months and the median Duration of Response was 7.7 months. The Clinical Benefit Rate was 45.4%. There was no meaningful difference in response rates in the various patient subgroups including patient age, onset of metastatic disease, number of previous therapies and the presence or absence of visceral metastases. The median PFS was 5.5 months and median OS was 13.0 months. The most common adverse reactions were, possibly severe neutropenia and diarrhea, fatigue, nausea, vomiting, alopecia and abdominal discomfort.

It was concluded that TRODELVY® was associated with durable Objective Responses in patients with heavily pretreated metastatic Triple Negative Breast Cancer. This unique Antibody Drug Conjugate may be of potential benefit for other Trop-2 expressing advanced epithelial solid tumors.
Sacituzumab Govitecan-hziy in Refractory Metastatic Triple-Negative Breast Cancer. Bardia A, Mayer IA, Vahdat LT, et al. N Engl J Med. 2019;380:741-751.

ELIQUIS® May Be Safer for the Treatment of Venous Thromboembolism Associated with Cancer

SUMMARY: The Center for Disease Control and Prevention (CDC) estimates that approximately 1-2 per 1000 individuals develop Deep Vein Thrombosis (DVT)/Pulmonary Embolism (PE) each year in the United States, resulting in 60,000-100,000 deaths. Venous ThromboEmbolism (VTE) is the third leading cause of cardiovascular mortality, after myocardial infarction and stroke. Ambulatory cancer patients initiating chemotherapy are at varying risk for Venous Thromboembolism (VTE), which in turn can have a substantial effect on health care costs, with negative impact on quality of life.

Approximately 20% of cancer patients develop VTE and there is a two-fold increase in the risk of recurrent thrombosis in patients with cancer, compared with those without cancer. The high risk of recurrent VTE, as well as bleeding in this patient group, makes anticoagulant treatment challenging. Treatment with parenteral Low Molecular Weight Heparin (LMWH) preparations is often recommended for this patient group, based on efficacy data. LMWH accelerates the inhibition by Antithrombin of activated Factor X, in the conversion of Prothrombin to Thrombin. Parenteral LMWH however can be inconvenient and expensive, leading to premature discontinuation of treatment.Anticoagulants

Direct Oral Anticoagulant agents have been proven to be as effective as COUMADIN® (Warfarin), a Vitamin K antagonist, for the treatment of VTE, and are associated with less frequent and less severe bleeding, and fewer drug interactions. The Direct Oral AntiCoagulants (DOACs) include PRADAXA® (Dabigatran), which is a direct Thrombin inhibitor and XARELTO® (Rivaroxaban), ELIQUIS® (Apixaban), SAVAYSA® (Edoxaban), BEVYXXA® (Betrixaban), which are Factor Xa inhibitors. Compared to COUMADIN®, the New Oral Anticoagulants have a rapid onset of action, wider therapeutic window, shorter half-lives (7-14 hours in healthy individuals), require no laboratory monitoring and have a fixed dosing schedule.

Three open-label, randomized, controlled trials have compared direct Factor Xa inhibitors with subcutaneous LMWH FRAGMIN® (Dalteparin). In the Hokusai VTE Cancer noninferiority trial, SAVAYSA® (Edoxaban)‎ when compared with FRAGMIN® was associated with a lower rate of recurrent VTE, but this was offset by a similar increase in the risk of major bleeding. In the SELECT-D trial, XARELTO® was associated with relatively low VTE recurrence in patients with cancer, but with higher clinically relevant non-major bleeding, compared with FRAGMIN®. In the ADAM VTE Trial, oral ELIQUIS® therapy was associated with very low rates of bleeding and significantly lower VTE recurrence compared to parenteral FRAGMIN®. The inconsistent results from these studies have been attributed to patient selection (cancer type, types of cancer therapies and prognosis), duration of treatment, and primary efficacy outcomes of these studies. SAVAYSA® and XARELTO® are often recommended as alternatives to LMWH in patients with cancer, although higher risk of clinically important bleeding has been reported with both these agents, particularly in patients with GI malignancies, including pancreatic cancer.

The Caravaggio trial is a multinational, randomized, open-label, noninferiority trial which was conducted to assess whether oral ELIQUIS® would be noninferior to subcutaneous FRAGMIN® (Dalteparin), a LMWH, for the prevention of recurrent VTE in patients with cancer, without increasing the risk of major bleeding. In this study, 1155 patients with cancer who had symptomatic or incidental acute proximal DVT or PE were randomly assigned to receive ELIQUIS® 10 mg orally twice daily for the first 7 days, followed by 5 mg orally twice daily (N=576) or FRAGMIN® 200 IU/kg administered subcutaneously once daily for the first month, followed by 150 IU/kg subcutaneous once daily (N=579). The demographic and clinical characteristics of the patients in both treatment groups were well balanced and advanced active cancers associated with high thromboembolic risk such as lung and colorectal cancers were well represented. This study included patients receiving a variety of cytotoxic and biologic therapies. Anticoagulant treatments were administered for 6 months. The Primary endpoint was objectively confirmed recurrent VTE during the trial period. The principal safety outcome was major bleeding.

The Primary endpoint of recurrent VTE occurred in 5.6% of patients in the ELIQUIS® group and in 7.9% of patients in the FRAGMIN® group (HR=0.63; P<0.001 for noninferiority). Major bleeding occurred in 3.8% of patients in the ELIQUIS® group and 4.0% of patients in the FRAGMIN® group (HR=0.82; P=0.60). Major GI bleeding occurred in 1.9% of patients in the ELIQUIS® group and in 1.7% of patients in the FRAGMIN® group and major non-gastrointestinal bleeding occurred in 1.9% and 2.2% of patients respectively. There were no fatal bleeding episodes noted in the ELIQUIS® group, whereas 2 patients had a fatal bleed in the FRAGMIN® group. These findings with regards to bleeding are in contrast to the results of previously published studies, which showed a higher incidence of bleeding with other Direct Oral AntiCoagulants, compared with FRAGMIN®, in a similar patient population.

It was concluded that in this study which included patients with predominantly advanced active cancer and acute symptomatic VTE, oral ELIQUIS® was noninferior to subcutaneous FRAGMIN® for the treatment of cancer-associated VTE, without an increased risk of major bleeding. The authors added that these findings may expand the proportion of patients with both cancer and VTE who would be eligible for treatment with ELIQUIS®, including patients with active gastrointestinal malignancies. It should be noted however that LMWH should still be preferred for patients who have undergone surgery involving the upper GI tract, as Direct Oral AntiCoagulants are absorbed in the stomach or proximal small bowel, as well as for those patients with bleeding or thrombocytopenia, recurrent VTE, CNS cancers, or those with severe renal impairment, and in the perioperative setting.
Apixaban for the Treatment of Venous Thromboembolism Associated with Cancer. Agnelli G, Becattini C, Meyer G, et al. for the Caravaggio Investigators. N Engl J Med 2020; 382:1599-1607