FDA Approves ALIQOPA®, a PI3K Inhibitor, for Follicular Lymphoma

SUMMARY: The FDA on September 14, 2017, granted accelerated approval to ALIQOPA® (Copanlisib) for the treatment of adult patients with relapsed Follicular Lymphoma, who have received at least two prior systemic therapies. The American Cancer Society estimates that in 2017, about 72,240 people will be diagnosed with Non Hodgkin Lymphoma (NHL) in the United States and about 20,140 individuals will die of this disease. Indolent NHLs are mature B cell lymphoproliferative disorders and include Follicular Lymphoma, Nodal Marginal Zone Lymphoma (NMZL), Extranodal Marginal Zone Lymphoma (ENMZL) of Mucosa-Associated Lymphoid Tissue (MALT), Splenic Marginal Zone Lymphoma (SMZL), LymphoPlasmacytic Lymphoma (LPL) and Small Lymphocytic Lymphoma (SLL). Follicular Lymphoma is the most indolent form and second most common form of all NHLs and they are a heterogeneous group of lymphoproliferative malignancies. Approximately 20% of all NHLs are Follicular Lymphomas. Advanced stage indolent NHL is not curable and as such, prolonging Progression Free Survival (PFS) and Overall Survival (OS), while maintaining Quality of Life, have been the goals of treatment intervention. Asymptomatic patients with indolent NHL are generally considered candidates for “watch and wait” approach whereas those with B symptoms (fever, night sweats, and weight loss), painful lymphadenopathy/splenomegaly, organ compromise and cytopenias are generally considered candidates for therapy.. Follicular Lymphoma International Prognostic Index (FLIPI) is of prognostic value and is used to help with treatment choices. The Ann Arbor classification divides FL into four stages. Patients with stages I and II have localized disease and those with stages III and IV have advanced disease. The World Health Organization (WHO) further classified FL based on histology into low grade (grades 1 and 2) and high grade (grade 3a) FLs. Grade 3b FL which demonstrates diffuse areas of involvement is designated as Diffuse Large B-cell Lymphoma (DLBCL) and is treated as such. Patients with advanced stage symptomatic Follicular Lymphoma are often treated with induction chemoimmunotherapy followed by maintenance RITUXAN® (Rituximab). This can result in a median PFS of 6-8 yrs and a median Overall Survival of 12-15 yrs. However, approximately 30% of the patients will relapse in 3 years and treatment options are limited for patients with relapses, after multiple treatments.

ALIQOPA® is a pan-class 1, PI3K inhibitor with inhibitory activity predominantly against PI3K-α and PI3K-δ Isoforms expressed in malignant B cells. The alpha isoform is broadly expressed and involved in insulin signaling and angiogenesis, as well as resistance mechanisms to lymphoma whereas the delta isoform is expressed by leukocytes and is involved in B-cell signaling, development, and survival. ALIQOPA® has been shown to induce tumor cell death by apoptosis and inhibition of proliferation of primary malignant B cell lines. ALIQOPA® also inhibits several key cell-signaling pathways, including B-cell receptor (BCR) signaling, CXCR12 (C-X-C chemokine receptor 12) mediated chemotaxis of malignant B cells, and NFÏ°B (Nuclear Factor Kappa B) signaling in lymphoma cell lines.

The approval of ALIQOPA® was based on data from the CHRONOS-1 trial, which is an open-label, single arm, multicenter, phase II study of patients with relapsed, Indolent or aggressive Non Hodgkin Lymphomas. This trial included patients with Follicular lymphoma (Grades 1-3a), Marginal Zone Lymphoma, Small Lymphocytic Lymphoma, and LymphoPlasmacytic Lymphoma /Waldenstrom Macroglobulinemia . Eligible patients had relapsed or refractory disease and had received at least two prior systemic therapies. The efficacy data leading to the FDA approval included 104 patients with Follicular B-cell Non Hodgkin Lymphoma. In this trial, patients received 0.8 mg/kg or 60 mg of ALIQOPA® by IV infusion on days 1, 8, and 15 of a 28-day treatment cycle, until disease progression or development of unacceptable toxicity. The median patient age was 63 yrs and all study patients had prior exposure to RITUXAN® and one or more alkylating agents, and 60% of the patients had disease that was refractory to the last regimen received. The Primary endpoint was Objective Response Rate after a minimum of 16 weeks of treatment. Secondary endpoints included Progression Free Survival, Duration of Response, Overall Survival, safety, and Quality of Life.

The Objective Response Rate was 58.7%, with an estimated median response duration of 12.2 months. The Complete Response rate was 14.4% and partial response rate was 44.3% and 33.7% has stable disease. The most common adverse reactions included nausea, hyperglycemia, diarrhea, fatigue, hypertension, cytopenias and lower respiratory tract infections. According to the authors, safety was manageable compared with the other PI3K inhibitor approved by the FDA, ZYDELIG® (Idelalisib), which targets only the δ-isoform and has warnings against fatal or severe colitis, intestinal perforation, hepatotoxicity, and pneumonitis The safety advantage with ALIQOPA® may be due to the dose scheduling or the IV mode of delivery.

It was concluded that ALIQOPA® has significant activity in patients with relapsed/refractory indolent B-cell lymphoma, and the safety was manageable, compared with other PI3K inhibitors. Two phase III trials are underway, using ALIQOPA® in combination with RITUXAN®. Dreyling M, Santoro A, Mollica L, et al. Copanlisib in patients with relapsed or refractory indolent B-cell lymphoma: Primary results of the pivotal CHRONOS-1 study. Presented at: 2017 AACR Annual Meeting; April 1-5, 2017; Washington, DC. Abstract CT149.

FDA Approves KEYTRUDA® for Advanced Gastric Cancer

SUMMARY: The FDA on September 22, 2017 granted accelerated approval to KEYTRUDA® (Pembrolizumab), for patients with recurrent locally advanced or metastatic, Gastric or GastroEsophageal junction adenocarcinoma, whose tumors express PD-L1, as determined by an FDA-approved test. Patients must have had disease progression on or after two or more prior systemic therapies, including Fluoropyrimidine and platinum-containing chemotherapy and, if appropriate, HER2/neu-targeted therapy. Cancers of the esophagus and stomach are among the most prevalent malignancies and are a major cause of cancer-related mortality. It is estimated that in 2016 GastroEsophageal Adenocarcinoma (GEA) accounted for 43,280 new cases in the United States. The incidence of these tumors has been on the rise in the past decade. Majority of the patients with GEA have advanced disease at the time of initial presentation and have limited therapeutic options with little or no chance for cure. 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.

KEYTRUDA® 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, thereby undoing PD-1 pathway-mediated inhibition of the immune response and unleashing the tumor-specific effector T cells. In previously reported Phase I study, KEYTRUDA® showed promising antitumor activity with manageable safety, in patients with previously treated advanced gastric cancer.

Based on these findings, the authors conducted a global, multicohort, Phase II study (KEYNOTE 059), in which patients with advanced gastric or gastroesophageal junction cancer received KEYTRUDA® 200 mg every 3 weeks for up to 2 years or until disease progression or unacceptable toxicity. Cohort 1 in this study enrolled 259 patients of whom 57% (N=148) had PD-L1 positive tumors and these tumors were either MicroSatellite Stable (MSS), or had undetermined MicroSatellite Instability (MSI) or MisMatch Repair (MMR) status. PD-L1 expression was evaluated by the PD-L1 IHC (22C3 antibody) and PD-L1 positivity was based on a Combined Positive Score (CPS) of 1 or more. CPS is determined by the number of PD-L1 staining cells (tumor cells, lymphocytes, macrophages) divided by total number of tumor cells evaluated, multiplied by 100. The median age was 62 years, 76% were men and over 50% of the patients received KEYTRUDA® as third line treatment or beyond. The median duration of follow up was 5.4 months. Primary end points included Objective Response Rate (ORR), safety, and tolerability.

In the group of patients with PD-L1 positive tumors, the ORR was 15.5% with 2% Complete Responses and 13.5% Partial Responses. Among the responding patients, the response duration ranged from 3-19 months, with 58% of the responding patients having response durations of 6 months or longer and 26% of the responding patients having response durations of 12 months or longer. In this cohort of 259 patients, 3% had tumors that were determined to be MSI-High. The ORR in this group was 57%, with response duration ranging from 5-14 months. In the PD-L1 negative patients however, the ORR was only 5.5%.

It was concluded that KEYTRUDA® demonstrated promising antitumor activity and durable responses in patients with advanced Gastric/GastroEsophageal Junction cancer, who had progressed on more than 2 lines of therapy, with higher Objective Response Rates noted in patients with PD-L1-positive tumors. KEYNOTE-059 cohort 1: Efficacy and safety of pembrolizumab (pembro) monotherapy in patients with previously treated advanced gastric cancer. Fuchs CS, Doi T, Jang RW-J, et al. J Clin Oncol. 2017;35 (suppl; abstr 4003).

FDA Approves NERLYNX® for Adjuvant Treatment of HER2 Positive Breast Cancer

The FDA on July 17, 2017 approved NERLYNX® (Neratinib) for the extended adjuvant treatment of adult patients with early stage HER2-overexpressed/amplified breast cancer, to follow adjuvant Trastuzumab (HERCEPTIN®)-based therapy. NERLYNX® is a potent, irreversible, oral Tyrosine Kinase Inhibitor, of HER1, HER2 and HER4 (pan-HER inhibitor). NERLYNX® is the first TKI approved by the FDA, shown to reduce the risk for disease recurrence, in patients with early stage HER2-positive breast cancer and demonstrated significantly improved 2-year invasive Disease Free Survival.

E-Cigarettes Can Increase the Risk of Bladder Cancer

SUMMARY: The American Cancer Society estimates that tobacco use is responsible for nearly 1 in 5 deaths in the United States and accounts for at least 30% of all cancer deaths. Smokeless tobacco products are a major source of cancer causing nitrosamines, and increase the risk of developing cancer of the oropharynx, esophagus, and pancreas. Cigarette smoke contains more than 7,000 chemicals, many of which are toxic and some linked to cancer. E-cigarettes or Electronic Nicotine Delivery Systems (ENDS) were first developed in China and introduced to the U.S. market in 2007. When a smoker inhales through the mouth piece of an e-cigarette, the air flow triggers a sensor that switches on a small lithium battery powered heater, which in turn vaporizes liquid nicotine along with PolyEthylene Glycol (PEG) present in a small cartridge. The PEG vapor looks like smoke. The potent liquid form of nicotine extracted from tobacco is tinctured with fragrant flavors such as chocolate, cherry and bubble gum, coloring substances, as well other chemicals and these e-liquids are powerful neurotoxins.

With the rapid growth of the e-cigarette industry and the evidence of potential dangers and risk to public health, particularly children, experts from the world's leading lung organizations were compelled to release a position statement on electronic cigarettes, specifically focusing on their potential adverse effects on human health and calling on government organizations to ban or restrict the use of e-cigarettes, until their impact on health is better understood. According to the National Youth Tobacco Survey, the use of e-cigarettes has tripled from 2013 to 2014 among middle school and high school students. Epidemiological data have shown that nicotine use is a gateway to the use of cocaine and marijuana and subsequent lifelong addiction. E-cigarettes and other Electronic Nicotine Delivery Systems (ENDS), unlike combustible cigarettes and many other tobacco products are not currently regulated by the U.S. Food and Drug Administration.

Smoking more than one pack of cigarettes a day has been associated with an increased risk of mortality among patients with bladder cancer. E-cigarettes have been advertised and promoted as a safer way of delivering the stimulating effects of tobacco smoke, without the harmful health risks. Contrary to this claim, over 90% of inhaled nicotine is excreted to urine. Nicotine can be nitrosatized in urothelial cells and then further metabolized into carcinogenic nitrosamines and formaldehyde, which in turn can induce DNA damage in the bladder mucosa. Additionally, these metabolites can also block DNA repair, increasing cancer risk.

The authors in this study compared the urine of e-cigarette users to that of nonsmokers, for known bladder carcinogens. There are five known bladder carcinogens that are either present in traditional cigarettes or common solvents believed to be used in some e-cigarette formulations, and they include (benz(a)anthracene, benzo(a)pyrene, 1-hydroxypyrene, o-toluidine and 2-naphthylamine. The limit of detection of these carcinogens in this study was 10-100 ng/ml. Urine samples were collected from 13 e-cigarette users and 10 non-smoking, non e-cigarette using controls. The mean age was 39 years and participants were predominantly male and had abstained completely from traditional cigarettes, for at least 6 months prior to specimen collection.

It was noted that the urine from 92% of e-cigarette users tested positive for two of the five carcinogenic compounds whereas none of the controls tested positive for these carcinogens. Interestingly, all study participants considered e-cigarettes were safe, when interviewed by the study investigators.

It was concluded that in this important pilot study, majority of the e-cigarette users had carcinogenic metabolites in the urine, which can contribute to the development of bladder cancer. This study underscores the importance both traditional and e-cigarette smoking cessation, for bladder cancer prevention. MP88-14 EVALUATION OF E-CIGARETTES USERS URINE FOR KNOWN BLADDER CARCINOGENS. Fuller T, Acharya A, Bhaskar G, et al. DOI: http://dx.doi.org/10.1016/j.juro.2017.02.2739

FDA Approves First Biosimilar for Cancer Treatment

SUMMARY: The FDA on Sept. 14, 2017 approved MVASI® (Bevacizumab-awwb) as a Biosimilar to AVASTIN® (Bevacizumab). Bevacizumab is a recombinant immunoglobulin G1 (IgG1) monoclonal antibody (mAb) that binds to Vascular Endothelial Growth Factor (VEGF) and inhibits the interaction of VEGF with its receptors, VEGF receptor-1 and VEGF receptor-2. This in turn inhibits establishment of new blood vessels that are essential for the maintenance and growth of solid tumors. MVASI® is the first Biosimilar approved in the U.S. for the treatment of cancer. A Biosimilar product is a biological product that is approved based on its high similarity to an already approved biological product (also known as reference product). Biological products are made from living organisms including humans, animals and microorganisms such as bacteria or yeast, and are manufactured through biotechnology, derived from natural sources or produced synthetically. Biological products have larger molecules with a complex structure, than conventional drugs (also known as small molecule drugs).

Unlike biological products, conventional drugs are made of pure chemical substances and their structures can be identified. A generic drug is a copy of brand name drug and has the same active ingredient and is the same as brand name drug in dosage form, safety and strength, route of administration, quality, performance characteristics and intended use. Therefore, brand name and the generic drugs are bioequivalent. The Affordable Care Act in 2010 created an abbreviated licensure pathway for biological products that are demonstrated to be “Biosimilar” to, or “interchangeable” with an FDA approved biological product (reference product). The Biosimilar must show that it has no clinically meaningful differences in terms of safety and effectiveness from the reference product. A Biosimilar product can only be approved by the FDA if it has the same mechanism of action, route of administration, dosage form and strength as the reference product, and only for the indications and conditions of use that have been approved for the reference product. Biosimilars are not as easy to manufacture as generics (copies of brand name drugs), because of the complexity of the structure of the biologic product and the process used to make a biologic product. The facilities where Biosimilars are manufactured must also meet the FDA standards.

MVASI® is approved for the treatment of patients with the following cancers:

• Metastatic Colorectal cancer, in combination with intravenous 5-Fluorouracil-based chemotherapy for first or second line treatment. MVASI® is not indicated for the adjuvant treatment of surgically resected Colorectal cancer.

• Metastatic Colorectal cancer, in combination with Fluoropyrimidine-Irinotecan or Fluoropyrmidine-Oxaliplatin-based chemotherapy for the second line treatment of patients who have progressed on a first-line Bevacizumab containing regimen.

• Non-squamous Non Small Cell Lung Cancer, in combination with Carboplatin and Paclitaxel for first line treatment of unresectable, locally advanced, recurrent or metastatic disease.

• Glioblastoma with progressive disease following prior therapy, based on improvement in Objective Response Rate. No data is available demonstrating improvement in disease-related symptoms or survival with Bevacizumab.

• Metastatic Renal cell carcinoma, in combination with Interferon alfa.

• Cervical cancer that is persistent, recurrent, or metastatic disease, in combination with Paclitaxel and Cisplatin or Paclitaxel and Topotecan.

The approval of MVASI® was based on two studies. In the first study, PharmacoKinetics (PK) of biosimilar MVASI® was compared with Bevacizumab, following a single infusion of 3 mg/kg. It was concluded that the PK data was similar between the Biosimilar, MVASI® and Bevacizumab. The second study is a randomized, double-blind, phase III trial, that evaluated the efficacy and safety of MVASI®, compared with Bevacizumab, in patients with non-squamous Non Small Cell Lung Cancer (NSCLC). Patients with non-squamous NSCLC, on first line chemotherapy with Carboplatin and TAXOL® (Paclitaxel), were randomized in a 1:1 ratio to receive either MVASI® (N=328) or Bevacizumab 15 mg/kg (N=314), as an IV infusion, every 3 weeks, for 6 cycles. The Objective Response Rate (ORR) was similar between the two treatment groups (39.0% for MVASI® and 41.7% for Bevacizumab) and these results were not statistically different. The Duration of Response was similar. Adverse events were comparable in the two treatment groups. This study demonstrated that MVASI® was clinically similar to Bevacizumab.

The FDA concluded that the approval of MVASI® was based on comparisons of extensive structural and functional product characterization, animal data, human PharmacoKinetic and pharmacodynamic data, clinical immunogenicity, between MVASI® and AVASTIN® (Bevacizumab), and it was noted that MVASI® is highly similar to AVASTIN® and that there are no clinically meaningful differences between the two products. Randomized, double-blind, phase 3 study evaluating efficacy and safety of ABP 215 compared with bevacizumab in patients with non-squamous NSCLC. Thatcher N, Thomas M, Paz-Ares L, et al. DOI: 10.1200/JCO.2016.34.15_suppl.9095 Journal of Clinical Oncology 34, no. 15_suppl (May 2016) 9095-9095.

KYMRIAH® (Tisagenlecleucel)

The FDA on August 30, 2017 granted regular approval to KYMRIAH®, for the treatment of patients up to age 25 years with B-cell precursor Acute Lymphoblastic Leukemia (ALL), that is refractory or in second or later relapse. KYMRIAH® is a product of Novartis Pharmaceuticals Corp.

FDA Approves First Biosimilar for Cancer Treatment

The FDA on Sept. 14, 2017 approved MYASI® (Bevacizumab-awwb) as a Biosimilar to AVASTIN® (Bevacizumab). MYASI® is the first Biosimilar approved in the U.S. for the treatment of cancer. A Biosimilar must show that it has no clinically meaningful differences in terms of safety and effectiveness from the already approved biological product (also known as reference product). A Biosimilar product can only be approved by the FDA if it has the same mechanism of action, route of administration, dosage form and strength as the reference product, and only for the indications and conditions of use, that have been approved for the reference product. The approval of MYASI® was based on comparisons of extensive structural and functional product characterization, animal data, human PharmacoKinetic and pharmacodynamic data, clinical immunogenicity, between MYASI® and AVASTIN® (Bevacizumab), and it was noted that MYASI® is highly similar to AVASTIN® and that there are no clinically meaningful differences between the two products.

Guideline for HER2 Testing in Gastroesophageal Adenocarcinoma

SUMMARY: It is estimated that in 2016 GastroEsophageal Adenocarcinoma (GEA) accounted for 43,280 new cases in the United States. Majority of the patients with GEA have advanced disease at the time of initial presentation and have limited therapeutic options with little or no chance for cure. 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. Approximately 7-38% of GEAs have amplification and/or overexpression of HER2. HER2 (ERBB2) is a proto-oncogene located on the long arm of chromosome 17 (17q12) that encodes a tyrosine kinase receptor, which upon dimerization and phosphorylation, initiates signaling pathways, that lead to cell division, proliferation, differentiation and anti-apoptosis signaling.

GastroEsophageal junction tumors have a higher incidence of overexpression of HER2 compared to gastric cancers. In the stomach, overexpression of HER2 varies with histologic type (intestinal-type having greater expression than diffuse-type) and differentiation (well and moderately differentiated tumors having greater expression than poorly differentiated tumors). Further, the heterogeneity of immunostaining is greater in GEA and the complete membrane staining required for positivity in breast cancer, is not common in GEA. Due to the high heterogeneity of a gastric cancer, it is appropriate to evaluate HER2 expression on biopsy specimens, especially in unresectable cases and reevaluate it on resected specimens, when available.

The NCCN (National Comprehensive Cancer Network) Clinical Practice Guidelines in Oncology for Gastric Cancer and Esophageal and Esophagogastric Junction Cancers, recommend assessment of HER2 overexpression using ImmunoHistoChemistry and/or gene amplification using FISH (Fluorescence In Situ Hybridization or another in situ hybridization method, in tumor samples from patients, with unresectable, locally advanced, recurrent, or metastatic GEA, for whom HERCEPTIN® may be potentially beneficial. Testing for HER2 is primarily performed on Formalin-Fixed and Paraffin-Embedded biopsy or resection tumor tissue from the primary or metastatic site. There are important differences in HER2 expression, scoring, and outcomes in GEA compared to breast carcinoma, and the need for HER2 guidelines (that include critical clinical and laboratory considerations) was therefore recognized. The CAP, American Society for Clinical Pathology, and ASCO convened an international expert panel to systematically review published documents and to develop an evidence-based guideline, to establish recommendations for HER2 testing in GastroEsophageal Adenocarcinoma.

HERCEPTIN® (Trastuzumab) is humanized monoclonal antibody that targets HER2. HERCEPTIN® is effective only in cancers where HER2 is overexpressed. In a previously published randomized phase III trial, HERCEPTIN® in combination with chemotherapy was shown to significantly improve Overall Survival compared with chemotherapy alone, in patients with HER2 positive advanced GEA.

The following are the Guideline Summary from the College of American Pathologists, American Society for Clinical Pathology, and American Society of Clinical Oncology

Guideline Questions

• What is the optimal testing algorithm for the assessment of Human Epithelial Growth Factor 2 (HER2) status in patients with GastroEsophageal Adenocarcinoma (GEA)?

• What strategies can help ensure optimal performance, interpretation, and reporting of established assays in patients with GEA?

Target Population: Patients with GEA.

Target Audience: Medical and surgical oncologists; oncology nurses and physician assistants; pathologists; general practitioners; and patients.

Key Points and Recommendations for Clinicians

Recommendation 1.1

For patients with advanced GEA who are potential candidates for HER2-targeted therapy, the treating clinician should request HER2 testing on tumor tissue.

Recommendation 1.2

Treating clinicians or pathologists should request HER2 testing on tumor tissue in the biopsy or resection specimens (primary or metastasis) preferably before the initiation of HERCEPTIN® therapy if such specimens are available and adequate. HER2 testing on fine-needle aspiration specimens (cell blocks) is an acceptable alternative.

Recommendation 1.3

Treating clinicians should offer combination chemotherapy and HER2-targeted therapy as the initial treatment of appropriate patients with HER2-positive tumors who have advanced GEA

Key Points and Recommendations for Pathologists

Recommendation 2.1

Laboratories/pathologists must specify the antibodies and probes used for the test and ensure that assays are appropriately validated for HER2 ImmunoHistoChemistry (IHC) and in situ hybridization (ISH) on GEA specimens.

Recommendation 2.2

When GEA HER2 status is being evaluated, laboratories/pathologists should perform/order IHC testing first, followed by ISH (In Situ Hybridization) when the IHC result is 2+ (equivocal). Positive (3+) or negative (0 or 1+) HER2 IHC results do not require further ISH testing.

Recommendation 2.3

Pathologists should use the Ruschoff/Hofmann method in scoring HER2 IHC and ISH results for GEA.

Recommendation 2.4

Pathologists should select the tissue block with the areas of lowest grade tumor morphology in biopsy and resection specimens. More than one tissue block may be selected if different morphologic patterns are present.

Recommendation 2.5

Laboratories should report HER2 test results in GEA specimens in accordance with the College of American Pathologists biomarker Template for Reporting Results of HER2 (ERBB2) Biomarker Testing of Specimens From Patients With Adenocarcinoma of the Stomach or Esophagogastric Junction.

Recommendation 2.6

Pathologists should identify areas of invasive adenocarcinoma and also mark areas with strongest intensity of HER2 expression by IHC in GEA specimens for subsequent ISH scoring when required.

Recommendation 2.7

Laboratories must incorporate GEA HER2 testing methods into their overall laboratory quality improvement program, establishing appropriate quality improvement monitors as needed to ensure consistent performance in all steps of the testing and reporting process. In particular, laboratories performing GEA HER2 testing should participate in a formal proficiency testing program, if available, or an alternative proficiency assurance activity.

Recommendation 2.8

There is insufficient evidence to recommend for or against genomic testing in patients with GEA at this time.

HER2 Testing and Clinical Decision Making in Gastroesophageal Adenocarcinoma: Guideline Summary From the College of American Pathologists, American Society for Clinical Pathology, and American Society of Clinical Oncology. Bartley AN, Washington MK, Ismaila N, et al. Journal of Oncology Practice 2017;13:53-57.

Maintenance Therapy with REVLIMID® Prolongs PFS in High Risk CLL

SUMMARY: The American Cancer Society estimates that approximately 20,110 new cases of Chronic Lymphocytic Leukemia (CLL) will be diagnosed in 2017 and approximately 4660 patients will die from the disease. CLL is a heterogeneous disease with a clinical course that is variable, with a very indolent course in some patients and some with aggressive disease and others somewhere in between. Both Binet and the Rai CLL staging systems developed in the 1970’s rely solely on physical examination and standard laboratory testing to predict survival. With the development of Interphase Fluorescent In Situ Hybridization (FISH) technique, which allows detection of genetic abnormalities in noncycling CLL cells, it has become clear that cytogenetic abnormalities are often seen in CLL patients and these genetic abnormalities in turn appear to be reliable predictors of disease progression, response to therapy and survival. Some of these cytogenetic abnormalities include del(13q), normal karyotype, trisomy(12), del(11q), del(17p), and they are associated with decreasing survival times, in that order. Another important prognostic factor is the rearrangement and somatic hypermutation of the variable region of the immunoglobulin heavy chain genes (IGHV), which is an independent predictor of outcome in CLL. Retrospective studies have suggested that patients with CLL whose leukemic cells unmutated IGHV gene demonstrated more aggressive disease and shorter survival time compared to those patients with somatic hypermutations in their IGHV genes (Mutated IGHV gene). Expression of two flow cytometry based biomarkers, CD38 (surface marker) and ZAP-70 (intracytoplasmic protein), have been associated with poor outcomes as well.

The rationale for maintenance treatment in the CLL patients is based on previously published studies showing that about 30% of patients with CLL did not substantially benefit from chemoimmunotherapy alone. These patients belong to the high risk group, as defined by poor cytogenetics, positive Minimal Residual Disease at the end of chemoimmunotherapy and unmutated IGHV gene status. The combined use of Genetic markers and Minimal Residual Disease (MRD) assessment can therefore identify patients with CLL, who have a poor outcome with first line chemoimmunotherapy.

Maintenance therapy with REVLIMID® (Lenalidomide)/Dexamethasone is considered standard of care for patients with multiple myeloma, regardless of transplantation and for newly diagnosed non-transplant candidates, as this intervention was found to improve Progression Free Survival significantly, with a favorable safety profile. Two phase III studies demonstrating the benefit of maintenance treatment with REVLIMID® in patients with CLL, were presented at the 2016 American Society of Hematology (ASH) Annual Meeting & Exposition. The first study (CLL M1 study) was conducted to demonstrate the value of maintenance treatment with REVLIMID® in patients with high risk CLL, following first line chemoimmunotherapy. For this study, 468 patients were screened, of whom 89 patients (N=89) were considered as high risk for disease progression, following 4 cycles of chemoimmunotherapy. This was based on the following factors: MRD levels of 10-2 or higher or MRD levels of 10-4 or higher to less than 10-2, combined with either an unmutated IGHV gene status, del(17p) or TP53 mutation at baseline.

Patients received investigator’s choice of chemoimmunotherapy regimens which included Fludarabine/Cyclophosphamide/Rituximab (FCR), Fludarabine and Rituximab (FR), Fludarabine and Cyclophosphamide (FC), or Bendamustine and Rituximab (BR). The most common regimens administered were Bendamustine and Rituximab followed by FCR. Approximately 78% of all patients reached Minimal Residual Disease negativity and these patients were ineligible for this trial. The 89 eligible patients were randomized in a 2:1 ratio to receive REVLIMID® maintenance (N=60) or Placebo (N=29). Treatment with REVLIMID® or Placebo was started at 5 mg daily for the first cycle, and was subsequently increased to the target dose of 15 mg by the 7th cycle. The median age of these patients was 64 years. At randomization, 37% of patients had a high and 63% had an intermediate MRD level, respectively. Based on their risk for thromboembolic events, patients received either low dose aspirin daily or appropriate prophylactic anticoagulants. The primary endpoint was Progression Free Survival (PFS).

After a median follow up of 17.7 months, the median PFS for REVLIMID® group has not been reached and was 13.3 months for the Placebo group. This meant a relative risk reduction for disease progression of 80% and this was highly statistically significant (P < 0.00001). There was no difference in Overall Survival at the time of this interim analysis. Adverse events such as neutropenia, diarrhea, skin disorders, etc. were more frequently noted with REVLIMID® maintenance treatment.

The authors concluded that REVLIMID® maintenance treatment for high risk CLL patients, after first line chemoimmunotherapy, significantly prolonged Progression Free Survival and this study confirmed the prognostic significance of the MRD based risk assessment.

The second study, CONTINUUM Trial, is a multicenter, randomized, double-blinded phase III study, designed to evaluate the efficacy and safety of REVLIMID® as maintenance therapy in previously treated CLL patients. In this study, 314 CLL patients who at least had a partial response (PR) to second line therapy were randomized 1:1 to receive either REVLIMID® 2.5 mg once daily on days 1-28 of the first 28 day cycle, or matching Placebo. If tolerated, REVLIMID® dose was then increased to 5 mg daily from cycle 2, and further increased to 10 mg daily at cycle 7 and thereafter. Co-primary endpoints were Progression Free Survival (PFS) and Overall Survival and secondary endpoints included Safety, Tumor response, Duration of Response and Health related Quality of Life measures.

At a median follow up of 31.5 months, the median PFS was significantly longer for the REVLIMID® group compared to the Placebo group (33.9 vs 9.2 months; HR=0.40; P<0.001). This benefit with REVLIMID® maintenance was maintained in all patient subgroups regardless of age, prior response to chemotherapy and number of poor prognostic factors.

The authors in this trial concluded that REVLIMID® maintenance resulted in a 60% reduction in the risk of disease progression in patients with CLL following second line treatment, and there was no meaningful difference in the Quality of Life between the two treatment groups.

Taken together, these two phase III studies suggest that REVLIMID® maintenance therapy following either first line or second line chemoimmunotherapy, significantly prolongs Progression Free Survival in patients with CLL. This strategy will however not be applicable to patients receiving upfront chemotherapy-free regimens such as BTK Inhibitor, IMBRUVICA® (Ibrutinib).

1. Lenalidomide maintenance after front line therapy substantially prolongs progression free survival in high risk CLL. Fink AM, Bahlo J, Sandra R, et al. 2016 ASH Annual Meeting. Abstract 229. Presented December 3, 2016.

2. Results of the phase 3 study of lenalidomide versus placebo as maintenance therapy following second-line treatment for patients with chronic lymphocytic leukemia. Foà R, Schuh A, Zaritskey A, et al. 2016 ASH Annual Meeting. Abstract 230. Presented December 3, 2016.