AVEO Oncology and EUSA Pharma Announce Updated Interim Results from Phase 2 Portion of the TiNivo Study in Renal Cell Carcinoma

On October 22, 2018 AVEO Oncology (NASDAQ:AVEO) and EUSA Pharma reported the presentation of updated interim results from the Phase 2 portion of the TiNivo study, a Phase 1b/2 multicenter trial of oral (PO) tivozanib (FOTIVDA) in combination with intravenous (IV) nivolumab (OPDIVO, Bristol-Myers Squibb), an immune checkpoint, or PD-1, inhibitor, for the treatment of advanced or metastatic renal cell carcinoma (Press release, AVEO, OCT 22, 2018, View Source [SID1234530044]). The results were presented today at European Society of Medical Oncology (ESMO) (Free ESMO Whitepaper) 2018 Annual Congress, in a poster presentation titled "TiNivo: Tivozanib Combined with Nivolumab: Safety and Efficacy in Patients with Metastatic Renal Cell Carcinoma (mRCC)" (Presentation 878P). A copy of the presentation is available at www.aveooncology.com or further information can be obtained via EUSA Pharma Medical Information.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

The Phase 1b/2 study has enrolled a total of 28 patients. The Phase 2 portion of the study (n=22) was designed to assess the safety, tolerability, and anti-tumor activity of the full dose and schedule of PO tivozanib (1.5 mg/QD for 21 days followed by a 7-day rest period), as established in the Phase 1b portion of the study (n=6), in combination with IV nivolumab (240 mg every 2 weeks). The combination was generally well tolerated. Treatment-related Grade 3/4 adverse events occurred in 60% of patients, the most common of which was hypertension.

Interim efficacy was assessed in all 25 patients treated with the full dose and schedule of PO tivozanib in combination with IV nivolumab and enrolled at least 4 months prior to the data cutoff date. Of these, 13 (52%) had received at least one prior systemic therapy. An objective response rate was observed in 56% of patients (complete responses + partial responses), including 4% of patients (n=1) achieving a complete response, and a disease control rate (complete response + partial response + stable disease) was observed in 96% of patients. At the time of data collection 52% (n=13), of patients remained on study. To date, a total of 72% of patients (n=18) had tumor shrinkage of ≥25%, with a majority of patients having disease control for ≥48 weeks.

"With high and durable tumor shrinkage rates for the combination of tivozanib and nivolumab, including a complete response, coupled with a favorable tolerability profile and nearly all patients having disease control, the TiNivo study continues to underscore a compelling rationale for using a high-specificity VEGF inhibitor as the TKI of choice in immuno-oncology combinations," Doctor Bernard Escudier, MD, ex-Chairman of the Genitourinary Oncology Committee, Gustave Roussy, and lead investigator of the study. "The ability to give a VEGF inhibitor and immuno-oncology agent both at full dose and strength could serve to deliver both improved outcomes and an improved patient experience. I look forward to better understanding tivozanib’s potential in immunotherapy combinations through a larger randomized study, which is currently being planned."

"There are multiple cancers where IO-TKI combinations have demonstrated potential, and the favorable tolerability and efficacy outcomes seen in the TiNivo study make further exploration of these indications a priority for AVEO," said Michael Bailey, president and chief executive officer of AVEO. "We continue to build out a clinical strategy for studying such combinations, and look forward to outlining our plans following reporting of topline data from our Phase 3 TIVO-3 study, which is expected in the mid-fourth quarter."

"The data arising from combination studies with checkpoint inhibitors demonstrate the considerable potential for tivozanib in metastatic RCC," said Lee Morley, Chief Executive Officer of EUSA Pharma. "EUSA continues to launch tivozanib across the EU in line with its EMA approval as monotherapy in the first line setting where its efficacy and favorable tolerability profile continue to provide benefits to patients, and we are excited by the prospect of further development of tivozanib as part of a future IO-TKI treatment option."

About Tivozanib (FOTIVDA)

Tivozanib (FOTIVDA) is an oral, once-daily, vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) discovered by Kyowa Hakko Kirin and approved for the treatment of adult patients with advanced renal cell carcinoma (RCC) in the European Union plus Norway and Iceland. It is a potent, selective and long half-life inhibitor of all three VEGF receptors and is designed to optimize VEGF blockade while minimizing off-target toxicities, potentially resulting in improved efficacy and minimal dose modifications.1,2 Tivozanib has been shown to significantly reduce regulatory T-cell production in preclinical models, enabling potentially enhanced activity when used in combination with immune modulating therapy.3 As part of a North American registration plan, tivozanib is currently being studied in the Phase 3 TIVO-3 trial, a randomized, controlled, multi-center, open-label study to compare tivozanib to sorafenib in subjects with refractory RCC. Tivozanib has been investigated in several tumors types, including renal cell, hepatocellular, colorectal and breast cancers.

Epizyme Reports Positive Data on Tazemetostat in Epithelioid Sarcoma from its Phase 2 Trial Cohort at ESMO

On October 22, 2018 Epizyme, Inc. (Nasdaq: EPZM), a clinical-stage company developing novel epigenetic therapies, reported positive interim data from the fully enrolled epithelioid sarcoma cohort of its ongoing Phase 2 study of its lead candidate tazemetostat, a potent, selective, orally available EZH2 inhibitor (Press release, Epizyme, OCT 22, 2018, View Source [SID1234530043]). The data were presented by the study’s lead investigator, Mrinal Gounder, M.D., attending physician, Sarcoma Medical Oncology and Early Drug Development Services, Memorial Sloan Kettering Cancer Center, during the European Society for Medical Oncology (ESMO) (Free ESMO Whitepaper) 2018 Congress in Munich, Germany.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

Interim data as of August 21, 2018 from the 62 patients enrolled show that tazemetostat treatment demonstrated clinically meaningful activity for patients with epithelioid sarcoma (ES). Oral, twice daily administration of tazemetostat resulted in durable objective responses and encouraging overall survival in both treatment-naive patients and patients who had been previously treated with an anticancer therapy. In addition, tazemetostat was generally well-tolerated.

"Epithelioid sarcoma is a rare and aggressive form of soft tissue sarcoma, with limited effective treatment options available. We often see patients diagnosed with advanced disease who have a very poor prognosis," said Dr. Gounder. "It is highly encouraging to see these updated efficacy and tolerability data with tazemetostat, and I believe this agent has the potential to be an important treatment option for patients with epithelioid sarcoma and their treating physicians."

"We are very pleased with the interim data from this cohort, which represents the largest prospective study of epithelioid sarcoma with any approved or investigational anticancer treatment to date. The data presented today further demonstrate the potential treatment opportunity we see with tazemetostat," said Rob Bazemore, president and chief executive officer of Epizyme. "We are one step closer to achieving our mission of bringing epigenetic treatments to people with cancer and other serious diseases. We look forward to working with investigators and regulators in an effort to bring to market the potential first therapy indicated for patients with epithelioid sarcoma."

Epithelioid Sarcoma Interim Efficacy Data
Epithelioid sarcoma is an ultra-rare and aggressive soft tissue sarcoma, characterized by a loss of the INI1 protein. It is most commonly diagnosed in young adults (18-40 years old) and is often fatal, with a median survival of less than one year in treatment-naive patients. Today, there is no treatment indicated specifically for epithelioid sarcoma.

The ES cohort completed enrollment in July of 2017 with 24 treatment-naive patients and 38 relapsed and/or refractory patients for a total of 62 epithelioid sarcoma patients. The primary endpoint of the study is objective response rate (ORR), comprised of complete and partial responses as measured by RECIST 1.1. Key secondary endpoints include duration of response, overall survival, disease control rate (DCR; comprised of confirmed objective responses for any duration or disease stabilization of 32 weeks or more) and safety.

Interim findings are summarized below, based on an August 21, 2018 data cut-off date.

Notably, since the data cut-off, one patient in the treatment-naive group who had stable disease subsequently achieved an objective response. This additional patient brings the total to six responders (~25%) in this treatment-naive group and nine responders (15%) in the overall population, to date, with several patients with stable disease remaining on treatment.

Tazemetostat Interim Safety Data
Tazemetostat has been generally well-tolerated and continues to demonstrate favorable safety in the Phase 2 study, with no discontinuations or deaths due to treatment-related adverse events (AEs) observed. The majority of treatment-emergent adverse events (TEAEs) were grade 1 or 2. Only 13 percent of patients experienced a grade 3 or higher treatment-related AE. Treatment-related AEs with an incidence of 10 percent or greater were fatigue (26%), nausea (26%), decreased appetite (16%), vomiting (16%), diarrhea and weight decrease (13%) and anemia (10%).

"We are pleased with the clinically meaningful and durable objective responses observed in this study, with a number of stable disease patients who are still on treatment and have the potential to achieve an objective response in the future," stated Dr. Shefali Agarwal, medical oncologist and chief medical officer of Epizyme. "These updated efficacy and safety data from the completed ES cohort suggest that tazemetostat may play an important role for patients in the future, particularly when considering the known limitations and challenges with current treatment options, and further bolster our confidence in our first planned NDA submission for tazemetostat. I’d like to thank the study investigators, medical staffs, and most importantly, the patients and caregivers, who have participated in this trial and supported the ongoing development of tazemetostat."

Conference Call Information
Epizyme management will host a conference call today at 8:30 am E.T. To participate in the conference call, please dial 877-844-6886 (domestic) or 970-315-0315 (international) and refer to conference ID 8780088. The webcast can be accessed in the Investor Relations section of the company’s website at www.epizyme.com. The replay of the webcast will be available in the investor section of the company’s website for 60 days.

About the Tazemetostat Clinical Trial Program
Tazemetostat, a potent, selective, orally available, first-in-class EZH2 inhibitor, is currently being studied as a monotherapy in ongoing Phase 2 programs in certain molecularly defined solid tumors, including epithelioid sarcoma and other INI1-negative tumors; follicular lymphoma (FL); and combination studies in diffuse large B-cell lymphoma (DLBCL) and non–small cell lung cancer (NSCLC).

amcure Presents First Clinical Data for Lead Compound AMC303 at ESMO 2018 Congress

On October 22, 2018 amcure, a biopharmaceutical company developing first-in-class cancer therapeutics, presented clinical trial data from its lead oncology drug candidate, AMC303, at the European Society for Medical Oncology (ESMO) (Free ESMO Whitepaper) Congress in Munich (Press release, amcure, OCT 22, 2018, View Source [SID1234530042]). The data presented on Sunday, 21 October in a proffered oral presentation demonstrated the favorable safety profile of AMC303. The company is currently conducting a Phase 1b expansion cohort and expects to publish updates from the study in 2019.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

AMC303 is a cyclic peptide targeting CD44v6, a key cell membrane protein in pathways of several receptor-tyrosine kinases, such as c-MET, VEGFR-2 and RON. This approach provides a potential novel mechanism for the treatment of patients with advanced and solid tumors that have already begun to spread throughout the body. Trial results presented at ESMO (Free ESMO Whitepaper) 2018 have shown the compound to be well-tolerated in 27 patients with a total of 11 different cancer types, with a favorable PK profile. No related serious adverse events (SAEs) were reported, and most frequently reported related events were infusion related reactions and hypersensitivity (grade 1-2, in 22% of patients), followed by nausea, diarrhea and fatigue.

"AMC303 was well tolerated in a heavily pretreated and diverse cancer patient population. The most related adverse events were transient and manageable. AMC303 has thus the potential of being a safe therapeutic option with a unique and additive mechanism of action," said Dr. Emiliano Calvo, MD, Lead Investigator of the trial at the Hospital Madrid Norte Sanchinarro and Director at the START Madrid-CIOCC Early Phase Clinical Drug Development program.

"These encouraging data support the continuation of the trial into its second part, targeting patients with a moderate to high expression of the target molecule CD44v6 and selected cancer types with a confirmed squamous cell histology. We look forward to updating the community on the progress of this trial and publish additional data sets as they emerge," added Klaus Dembowsky, CEO of amcure GmbH.

The trial, conducted in Belgium and Spain, is designed to assess the safety, tolerability and pharmacokinetics of multiple and increasing doses of AMC303 as monotherapy in patients with advanced metastatic malignant solid tumors of epithelial origin. In addition, the study includes a comprehensive biomarker program. The study was designed to include a broad variety of tumor types in the first part of the study irrespective of the target expression and a tumor type-specific expansion cohort at the recommended dose for a subsequent Phase 2 study. With the expansion cohort, amcure focuses its patient selection on patients with a moderate to high expression of the target molecule CD44v6 in four specific tumor types of squamous tumors: head and neck squamous cell carcinoma (HNSCC), squamous non-small-cell lung carcinoma (NSCLC), esophageal and cervical tumors.

For more information on the trial please visit View Source

About AMC303
amcure’s lead compound, AMC303, is being developed as a potential treatment for patients with advanced and metastatic epithelial tumors, e.g. pancreatic cancer, head and neck cancer, gastric cancer, colorectal cancer, breast cancer and lung cancer. AMC303 has a high specificity for inhibiting CD44v6, a co-receptor required for signaling through multiple cellular pathways (c-Met, VEGFR-2, RON) involved in tumor growth, angiogenesis and the development and regression of metastases. AMC303 has demonstrated strong effects in various in vitro and in vivo assays.

Novartis COMBI-AD study of Tafinlar® + Mekinist® continues to demonstrate relapse free survival benefit in patients with BRAF V600-mutant stage III melanoma

On October 22, 2018 Novartis reported a new data analysis of COMBI-AD, a phase III multi-center study evaluating Tafinlar (dabrafenib) in combination with Mekinist (trametinib) in stage III adjuvant resected BRAF V600-mutant melanoma (Press release, Novartis, OCT 22, 2018, https://www.novartis.com/news/media-releases/novartis-combi-ad-study-tafinlar-mekinist-continues-demonstrate-relapse-free-survival-benefit-patients-braf-v600-mutant-stage-iii-melanoma [SID1234530040]). With extended study follow up, Tafinlar in combination with Mekinist continued to show more than 50% risk reduction in relapse free survival (RFS) versus placebo in patients with resected BRAF V600-mutant stage III melanoma. The updated COMBI-AD data was also used to generate a statistical cure-rate model that estimated the fraction of patients who may not relapse. The cure rate was 54% (95% CI, 49%-59%) in the Tafinlar + Mekinist arm compared to 37% (95% CI, 32%-42%) in the placebo arm[1]. These data were presented at the European Society of Medical Oncology (ESMO) (Free ESMO Whitepaper) in Munich, Germany (Abstract #LBA43) today and simultaneously published in The Journal of Clinical Oncology.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

A separate biomarker analysis was conducted to identify predictors of clinical outcome and treatment response. These analyses showed that subgroups of patients at a higher risk of relapse could be defined based on specific immune gene expression signatures (GES) and tumor mutation burden (TMB). Exploratory analysis of RFS in the treatment vs. placebo arms in all TMB/immune GES subgroups suggested that specific subgroups may have a greater RFS benefit, but the predictive value of TMB and immune GES warrants further validation in a prospective study[2].

"In addition to confirming prior relapse free survival results in the adjuvant setting, this biomarker analysis of COMBI-AD provides important information about the prognostic and potentially predictive value of TMB and immune gene expression signatures in resected BRAF V600-mutant melanoma patients," said Georgina Long, BSc., PhD, MBBS, FRACP, Medical Oncologist, Melanoma Institute Australia, The University of Sydney.

In the phase III COMBI-AD global study, at median follow-ups of 44 months (Tafinlar + Mekinist) and 42 months (placebo), the three- and four-year RFS rates were 59% ([95% CI, 0.55-0.64]) and 54% ([95% CI, 0.49-0.59]) in the Tafinlar + Mekinist arm and 40% ([95% CI, 0.35-0.45]) and 38% ([95% CI, 34%-44%]) in the placebo arm, respectively (HR, 0.49 [95% CI, 0.40-0.59]). RFS was also analyzed by subgroups defined by baseline disease stage by the American Joint Committee on Cancer 7th and 8th editions and included nodal metastatic burden, and ulceration status. A cure rate model estimated that the fraction of patients who may not relapse was 54% (95% CI, 49%-59%) in the Tafinlar + Mekinist arm compared with 37% (95% CI, 32%-42%) in the placebo arm[1]. The fraction of patients remaining relapse free long term was estimated using a Weibull mixture cure-rate model. No updated safety analysis was performed as all patients have completed treatment at the time of the updated RFS analysis[1].

"The data generated from the COMBI-AD study have the ability to transform treatment decisions for patients with BRAF V600 melanoma," said Samit Hirawat, MD, Head, Novartis Oncology Global Drug Development. "Not only do the results from the extended analysis continue to provide confirmation of the long-term benefit with adjuvant Tafinlar and Mekinist, but the comprehensive biomarker analysis of the largest adjuvant dataset to date highlight important prognostic information to identify patients at higher risk of relapse."

The BRAF gene belongs to a class of genes known as oncogenes and provides instructions for making a protein that helps transmit chemical signals from outside the cell to the cell’s nucleus. This protein is part of a signaling pathway known as the RAS/MAPK pathway, which controls several important cell functions. Specifically, the RAS/MAPK pathway regulates the growth and division (proliferation) of cells, the process by which cells mature to carry out specific functions (differentiation), cell movement (migration) and the self-destruction of cells (apoptosis). Chemical signaling through this pathway is essential for normal development before birth. When mutated, oncogenes have the potential to cause normal cells to become cancerous. During cancer treatment, targeted therapies may inhibit the mutation from occurring, thus slowing the growth of the cancer tumor[3].

About COMBI-AD
The COMBI-AD study evaluated Tafinlar + Mekinist among patients with stage III, BRAF V600-mutant melanoma without prior anticancer therapy, randomized within 12 weeks of complete surgical resection. Patients received the Tafinlar (150 mg BID) + Mekinist (2 mg QD) combination (n = 438) or matching placebos (n = 432). In the initial primary analysis, and after a median follow-up of 2.8 years, the primary endpoint was met in that the combination therapy significantly reduced the risk of disease recurrence or death by 53% vs. placebo (HR: 0.47 [95% CI: 0.39-0.58]; median not yet reached vs. 16.6 months, respectively; p<0.001). The combination treatment group also saw an improvement in a key secondary endpoint of OS (HR: 0.57 [95% CI: 0.42-0.79] p=0.0006, which did not cross the predefined interim analysis boundary of p=0.000019 to claim statistical significance).

Other secondary endpoints in the initial primary analysis where the combination demonstrated a clinically meaningful benefit included distant metastasis-free survival (DMFS) (HR: 0.51 [95% CI: 0.40-0.65]), and freedom from relapse (FFR) (HR: 0.47 [95% CI: 0.39-0.57])[4]. In a separate analysis, Tafinlar + Mekinist also demonstrated benefit regardless of baseline factors, including disease stage, nodal metastatic burden, and ulceration[5]. Adverse events (AEs) were consistent with other Tafinlar + Mekinist studies and no new safety signals were reported. Of patients treated with the combination, 97% experienced an AE, with 41% having grade 3/4 AEs and 26% having AEs leading to treatment discontinuation (vs. 88%, 14%, and 3%, respectively, with placebo)[4].

Based on updated data with median follow up of 44 months (Tafinlar + Mekinist) and 42 months (matching placebo), the 3- and 4- year relapse-survival benefit maintained at 59% (95% CI, 55%-64%) and 54% (95% CI, 49%-59%) in the dabrafenib plus trametinib arm and 40% (95% CI, 35%-45%) and 38% (95% CI, 34%-44%) in the placebo arm, respectively (HR, 0.49 [95% CI, 0.40-0.59]). The relapse-free survival benefit among the combination arm was observed across all patient subgroups, including stage III A, B and C. The estimated one-year, two-year, three-year, and four-year RFS were consistently higher than placebo (one year: 88% vs. 56%; two year: 67% vs. 44%; three year: 59% vs. 40%; four year: 54% vs. 38%). The combination treatment group also saw an improvement in the secondary endpoint of distant metastasis-free survival (DMFS) (HR: 0.53 [95% CI: 0.42-0.67]). No new safety analysis was performed[1].

About Melanoma
There are about 280,000 new diagnoses of melanoma (stages 0-IV) worldwide each year[6], approximately half of which have BRAF mutations[3]. Biomarker tests can determine whether a tumor has a BRAF mutation[7].

Melanoma is staged by how far it has metastasized. In stage III melanoma, tumors have spread to the regional lymph nodes, presenting a higher risk of recurrence or metastases[8]. Patients who receive surgical treatment for Stage III melanoma may have a high risk of recurrence because melanoma cells can remain in the body after surgery; almost half (44%) of patients receiving placebo per the COMBI-AD study had a recurrence of disease within the first year[4],[9]. Adjuvant therapy is additional treatment given after surgical resection, and may be recommended for patients with high-risk melanoma to help reduce the risk of melanoma returning[9].

About Tafinlar + Mekinist
Tafinlar + Mekinist target different kinases within the serine/threonine kinase family-BRAF and MEK1/2, respectively-in the RAS/RAF/MEK/ERK pathway, which is implicated in melanoma and NSCLC, among other cancers. When Tafinlar is used with Mekinist, the combination has been shown to slow tumor growth more than either drug alone.

Tafinlar + Mekinist have been investigated for the treatment of a variety of cancers as part of an ongoing clinical trial program. Tafinlar + Mekinist are approved in more than 60 countries, for uses including:

as monotherapy and in combination for the treatment of subjects with unresectable or metastatic melanoma with a BRAFV600 mutation
in combination for the adjuvant treatment of patients with Stage III melanoma with a BRAFV600 mutation, following complete resection
in combination for the treatment of patients with advanced NSCLC with a BRAFV600 mutation
in combination for the treatment of patients with locally advanced or metastatic ATC with a BRAFV600 mutation
Approved indications vary worldwide. Please refer to local labeling for indication language in a particular country.

Tafinlar + Mekinist Combination Important Safety Information
Tafinlar and Mekinist, in combination, may cause serious side effects such as the risk of new cancers, including both skin cancer and nonskin cancer. Patients should be advised to contact their health care provider immediately for a new wart, skin sore, or bump that bleeds or does not heal, or a change in the size or color of a mole.

When Tafinlar is used in combination with Mekinist, it can cause serious bleeding problems, especially in the brain or stomach, that can lead to death. Patients should be advised to call their health care provider and get medical help right away if they have any signs of bleeding, including headaches, dizziness, or feel weak, cough up blood or blood clots, vomit blood or their vomit looks like "coffee grounds," or red or black stools that look like tar.

Mekinist, alone or in combination with Tafinlar, can cause inflammation of the intestines or tears in the stomach or intestines that can lead to death. Patients should report to their health care provider immediately if they have any of the following symptoms: bleeding, diarrhea (loose stools) or more bowel movements than usual, stomach-area (abdomen) pain or tenderness, fever, or nausea.

Tafinlar, in combination with Mekinist, can cause blood clots in the arms or legs, which can travel to the lungs and can lead to death. Patients should be advised to get medical help right away if they have the following symptoms: chest pain, sudden shortness of breath or trouble breathing, pain in their legs with or without swelling, swelling in their arms or legs, or a cool or pale arm or leg.

The combination of Tafinlar and Mekinist can cause heart problems, including heart failure. A patient’s heart function should be checked before and during treatment. Patients should be advised to call their health care provider right away if they have any of the following signs and symptoms of a heart problem: feeling like their heart is pounding or racing, shortness of breath, swelling of their ankles and feet, or feeling lightheaded.

Tafinlar, in combination with Mekinist, can cause severe eye problems that can lead to blindness. Patients should be advised to call their health care provider right away if they get: blurred vision, loss of vision, or other vision changes, seeing color dots, halo (seeing blurred outline around objects), eye pain, swelling, or redness.

Tafinlar, in combination with Mekinist, can cause lung or breathing problems. Patients should be advised to tell their health care provider if they have new or worsening symptoms of lung or breathing problems, including shortness of breath or cough.

Fever is common during treatment with Tafinlar in combination with Mekinist, but may also be serious. In some cases, chills or shaking chills, too much fluid loss (dehydration), low blood pressure, dizziness, or kidney problems may happen with the fever. Patients should be advised to call their health care provider right away if they get a fever.

Rash and other skin reactions are common side effects of Tafinlar in combination with Mekinist. In some cases, these rashes and other skin reactions can be severe or serious, and may need to be treated in a hospital. Patients should be advised to call their health care provider if they get any of the following symptoms: skin rash that bothers them or does not go away, acne, redness, swelling, peeling, or tenderness of hands or feet, or skin redness.

Some people may develop high blood sugar or worsening diabetes during treatment with Tafinlar in combination with Mekinist. For patients who are diabetic, their health care provider should check their blood sugar levels closely during treatment. Their diabetes medicine may need to be changed. Patients should be advised to tell their health care provider if they have increased thirst, urinate more often than normal, or produce an increased amount of urine.

Tafinlar, in combination with Mekinist, may cause healthy red blood cells to break down too early in people with glucose-6-phosphate dehydrogenase deficiency. This may lead to a type of anemia called hemolytic anemia, where the body does not have enough healthy red blood cells. Patients should be advised to tell their health care provider if they have yellow skin (jaundice), weakness or dizziness, or shortness of breath.

Tafinlar, in combination with Mekinist, can cause new or worsening high blood pressure (hypertension). A patient’s blood pressure should be checked during treatment. Patients should be advised to tell their health care provider if they develop high blood pressure, their blood pressure worsens, or if they have severe headache, lightheadedness, blurry vision, or dizziness.

The most common side effects of Tafinlar, in combination with Mekinist, include fever, rash, nausea, fatigue, headache, chills, diarrhea, vomiting, high blood pressure (hypertension), joint aches, muscle aches, swelling of the face, arms, or legs, and cough.

Genentech’s Tecentriq Plus Chemotherapy (Carboplatin and Abraxane) as an Initial Treatment Helped People With Advanced Non-Squamous Non-Small Cell Lung Cancer Live Significantly Longer Compared to Chemotherapy Alone

On October 22, 2018 Genentech, a member of the Roche Group (SIX: RO, ROG; OTCQX: RHHBY), reported positive results from the Phase III IMpower130 study of Tecentriq (atezolizumab) plus chemotherapy (carboplatin and Abraxane [albumin-bound paclitaxel; nab-paclitaxel]) for the initial (first-line) treatment of people with previously untreated metastatic non-squamous non-small cell lung cancer (NSCLC) (Press release, Genentech, OCT 22, 2018, View Source [SID1234530039]). The analysis showed that Tecentriq plus chemotherapy helped people live significantly longer compared to chemotherapy alone (median overall survival [OS] = 18.6 versus 13.9 months; hazard ratio [HR] = 0.79; 95 percent CI: 0.64–0.98; p=0.033) in the intention-to-treat wild-type (ITT-WT) population. The Tecentriq-based combination also significantly reduced the risk of disease worsening or death (progression-free survival; PFS) compared to chemotherapy alone (median PFS=7.0 versus 5.5 months; HR=0.64; 95 percent CI: 0.54–0.77; p<0.0001) in the ITT-WT population. Safety for the Tecentriq plus chemotherapy combination appeared consistent with the known safety profile of the individual medicines, and no new safety signals were identified with the combination.

Schedule your 30 min Free 1stOncology Demo!
Discover why more than 1,500 members use 1stOncology™ to excel in:

Early/Late Stage Pipeline Development - Target Scouting - Clinical Biomarkers - Indication Selection & Expansion - BD&L Contacts - Conference Reports - Combinatorial Drug Settings - Companion Diagnostics - Drug Repositioning - First-in-class Analysis - Competitive Analysis - Deals & Licensing

                  Schedule Your 30 min Free Demo!

"Initial treatment with this Tecentriq-based combination provided a significant survival benefit for people with non-squamous non-small cell lung cancer, the most common form of lung cancer," said Sandra Horning, M.D., chief medical officer and head of Global Product Development. "Lung cancer is a complex disease and this combination could offer a new potential treatment option. We will work with global health authorities to bring this regimen to people living with this disease as soon as possible."

The data will be presented at the European Society for Medical Oncology (ESMO) (Free ESMO Whitepaper) 2018 Congress on October 22, 2018 from 9:15 – 9:30 a.m. CEST (Abstract LBA53; Hall A1 – Room 17).

About the IMpower130 study

IMpower130 is a Phase III, multicenter, open-label, randomized study evaluating the efficacy and safety of Tecentriq in combination with carboplatin and nab-paclitaxel versus chemotherapy (carboplatin and nab-paclitaxel) alone for chemotherapy-naïve patients with stage IV non-squamous NSCLC. The study enrolled 723 people who were randomized (2:1) to receive:

Tecentriq plus carboplatin and nab-paclitaxel (Arm A), or
Carboplatin and nab-paclitaxel (Arm B, control arm)
During the treatment-induction phase, people in Arm A received Tecentriq and carboplatin on day 1 of each 21-day cycle, and nab-paclitaxel on days 1, 8 and 15 of each 21-day cycle for 4 or 6 cycles or until loss of clinical benefit, whichever occurs first. People received Tecentriq during the maintenance treatment phase until loss of clinical benefit was observed.

During the treatment-induction phase, people in Arm B received carboplatin on day 1 and nab-paclitaxel on days 1, 8 and 15 of each 21-day cycle for 4 or 6 cycles or until disease progression, whichever occurs first. People received best supportive care during the maintenance treatment phase. Switch maintenance to pemetrexed was also permitted. People who were consented prior to a protocol revision were given the option to crossover to receive Tecentriq as monotherapy until disease progression.

The co-primary endpoints were:

PFS as determined by the investigator using RECIST v1.1 in the ITT-WT population
OS in the ITT-WT population
IMpower130 met its co-primary endpoints of OS and PFS.

A summary of the results are included below:

Arm A (Tecentriq plus chemotherapy) vs Arm B (chemotherapy) in ITT-WT


Arm A
n=451


Arm B
n=228

Median OS, months (95% CI)


18.6
(16.0–21.2)


13.9
(12.0–18.7)

HR (95% CI); P value 0.79 (0.64, 0.98); p=0.033
1-year OS, % (95% CI) 63.1 (58.59–67.66) 55.5 (48.89–62.17)
Confirmed ORR, % (95% CI)
49.2 (44.49–53.96)


31.9 (25.84–38.36)

Median DoR, months (95% CI)


8.4
(6.9–11.8)


6.1
(5.5–7.9)

Median PFS (95% CI), months


7.0
(6.2–7.3)


5.5
(4.4–5.9)

HR (95% CI); P value 0.64 (0.54, 0.77); p<0.0001
1-year PFS rate, % (95% CI)


29.1
(24.83–33.44)


14.1
(9.37–18.76)

CI, confidence interval; DoR, duration of response; HR, hazard ratio; ORR, objective response rate; PFS, progression-free survival

Safety for the Tecentriq and chemotherapy combination appeared consistent with the known safety profile of the individual medicines, and no new safety signals were identified with the combination. Grade 3-4 treatment-related adverse events (AEs) were reported in 73.2 percent of people receiving Tecentriq plus chemotherapy compared to 60.3 percent of people receiving chemotherapy alone. The most common Grade 3-4 AEs in people receiving Tecentriq plus chemotherapy were: an abnormal low count of a certain type of white blood cell (neutropenia, 32.1 percent), a decrease in red blood cells (anemia, 29.2 percent) and a decreased neutrophil count (12.1 percent).

About lung cancer

According to the American Cancer Society, it is estimated that more than 234,000 Americans will be diagnosed with lung cancer in 2018, and NSCLC accounts for 85 percent of all lung cancers. It is estimated that approximately 60 percent of lung cancer diagnoses in the United States are made when the disease is in the advanced stages.

About Tecentriq (atezolizumab)

Tecentriq is a monoclonal antibody designed to bind with a protein called PD-L1. Tecentriq is designed to bind to PD-L1 expressed on tumor cells and tumor-infiltrating immune cells, blocking its interactions with both PD-1 and B7.1 receptors. By inhibiting PD-L1, Tecentriq may enable the re-activation of T cells. Tecentriq may also affect normal cells.

Abraxane is a registered trademark of Abraxis Bioscience, LLC, a wholly owned subsidiary of Celgene Corporation.

Tecentriq U.S. Indication (pronounced ‘tē-SEN-trik’)

Tecentriq is a prescription medicine used to treat:

A type of bladder and urinary tract cancer called urothelial carcinoma.

Tecentriq may be used when your bladder cancer:
has spread or cannot be removed by surgery, and if you have any one of the following conditions:
you are not able to take chemotherapy that contains a medicine called cisplatin, and your doctor has tested your cancer and found high levels of a specific protein on your cancer called programmed death-ligand 1 (PD-L1), as determined by an FDA-approved test, or
you are not able to take chemotherapy that contains any platinum regardless of PD-L1 status on your cancer, or
you have tried chemotherapy that contains platinum, and it did not work or is no longer working
The approval of Tecentriq in these patients is based on a study that measured response rate and duration of response. There is an ongoing study to confirm clinical benefit.

A type of lung cancer called non-small cell lung cancer (NSCLC).

Tecentriq may be used when your lung cancer:
has spread or grown, and
you have tried chemotherapy that contains platinum, and it did not work or is no longer working
If your tumor has an abnormal EGFR or ALK gene, you should have also tried an FDA-approved therapy for tumors with these abnormal genes, and it did not work or is no longer working.

It is not known if Tecentriq is safe and effective in children.

Important Safety Information

What is the most important information about Tecentriq?

Tecentriq can cause the immune system to attack normal organs and tissues and can affect the way they work. These problems can sometimes become serious or life threatening and can lead to death.

Patients should call or see their healthcare provider right away if they get any symptoms of the following problems or these symptoms get worse.

Tecentriq can cause serious side effects, including:

Lung problems (pneumonitis)–signs and symptoms may include new or worsening cough, shortness of breath, and chest pain
Liver problems (hepatitis)–signs and symptoms of hepatitis may include yellowing of the skin or the whites of the eyes, severe nausea or vomiting, pain on the right side of the stomach area (abdomen), drowsiness, dark urine (tea colored), bleeding or bruising more easily than normal, and feeling less hungry than usual
Intestinal problems (colitis)–signs and symptoms of colitis may include diarrhea (loose stools) or more bowel movements than usual, blood or mucous in the stools or dark, tarry, sticky stools, and severe stomach area (abdomen) pain or tenderness
Hormone gland problems (especially the thyroid, adrenal glands, pancreas, and pituitary)–signs and symptoms that the hormone glands are not working properly may include headaches that will not go away or unusual headaches, extreme tiredness, weight gain or weight loss, dizziness or fainting, feeling more hungry or thirsty than usual, hair loss, changes in mood or behavior (such as decreased sex drive, irritability, or forgetfulness), feeling cold, constipation, the voice gets deeper, urinating more often than usual, nausea or vomiting, and stomach area (abdomen) pain
Problems in other organs–signs and symptoms may include severe muscle weakness, numbness or tingling in hands or feet, confusion, blurry vision, double vision, or other vision problems, changes in mood or behavior, extreme sensitivity to light, neck stiffness, eye pain or redness, skin blisters or peeling, chest pain, irregular heartbeat, shortness of breath, or swelling of the ankles
Severe infections–signs and symptoms of infection may include fever, cough, flu-like symptoms, pain when urinating, and frequent urination or back pain
Severe infusion reactions–signs and symptoms of infusion reactions may include chills or shaking, itching or rash, flushing, shortness of breath or wheezing, swelling of the face or lips, dizziness, fever, feeling like passing out, and back or neck pain
Getting medical treatment right away may help keep these problems from becoming more serious. A healthcare provider may treat patients with corticosteroid or hormone replacement medicines. A healthcare provider may delay or completely stop treatment with Tecentriq if patients have severe side effects.

Before receiving Tecentriq, patients should tell their healthcare provider about all of their medical conditions, including if they:

have immune system problems (such as Crohn’s disease, ulcerative colitis, or lupus); have had an organ transplant; have lung or breathing problems; have liver problems; have a condition that affects the nervous system (such as myasthenia gravis or Guillain-Barre syndrome); or are being treated for an infection
are pregnant or plan to become pregnant. Tecentriq can harm an unborn baby. Patients should tell their healthcare provider right away if they become pregnant or think they may be pregnant during treatment with Tecentriq. If patients are able to become pregnant:
A healthcare provider should do a pregnancy test before they start treatment with Tecentriq.
They should use an effective method of birth control during their treatment and for at least 5 months after the last dose of Tecentriq.
are breastfeeding or plan to breastfeed. It is not known if Tecentriq passes into the breast milk. Do not breastfeed during treatment and for at least 5 months after the last dose of Tecentriq
Patients should tell their healthcare provider about all the medicines they take, including prescription and over-the-counter medicines, vitamins, and herbal supplements.

The most common side effects of Tecentriq in people with urothelial carcinoma include:

feeling tired
decreased appetite
nausea
constipation
urinary tract infection
diarrhea
fever
The most common side effects of Tecentriq in people with non-small cell lung cancer include:

feeling tired
decreased appetite
muscle pain
cough
shortness of breath
Tecentriq may cause fertility problems in females, which may affect the ability to have children. Patients should talk to their healthcare provider if they have concerns about fertility.

These are not all the possible side effects of Tecentriq. Patients should ask their healthcare provider or pharmacist for more information. Patients should call their doctor for medical advice about side effects.