ImmunoGen Announces Conference Call to Discuss Its Second Quarter 2018 Operating Results

On July 12, 2018 ImmunoGen, Inc., (Nasdaq: IMGN), a leader in the expanding field of antibody-drug conjugates (ADCs) for the treatment of cancer, reported that Company will host a conference call at 8:00 a.m. ET on Friday, July 27, 2018 to discuss its second quarter operating results (Press release, ImmunoGen, JUL 12, 2018, View Source [SID1234527673]). Management will also provide a brief update on the business.

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!

Conference Call Information
To access the live call by phone, dial 719-785-1753; the conference ID is 2275763. The call may also be accessed through the Investors section of the Company’s website, www.immunogen.com. Following the webcast, a replay of the call will be available at the same location through August 10, 2018.

Genmab Enters Strategic Partnership with Immatics to Discover and Develop Next Generation Bispecific Cancer Immunotherapies

On July 12, 2018 Genmab A/S (Nasdaq Copenhagen: GEN) reported that it has entered into a research collaboration and exclusive license agreement with privately owned Immatics Biotechnologies GmbH (Immatics), to discover and develop next-generation bispecific immunotherapies to target multiple cancer indications (Press release, Genmab, JUL 12, 2018, View Source [SID1234527670]). The deal strengthens Genmab’s position in immuno-oncology by combining Genmab’s proprietary technologies and antibody know-how with Immatics’ XPRESIDENT targets and T-cell receptor (TCR) capabilities. Genmab will receive an exclusive license to three proprietary targets from Immatics, with an option to license up to two additional targets at predetermined economics. The companies will conduct joint research, funded by Genmab, on multiple antibody and/or TCR-based bispecific therapeutic product concepts.

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!

Genmab may elect to progress any resulting product candidates, and will be responsible for development, manufacturing and worldwide commercialization. For any products that are commercialized by Genmab, Immatics will have an option to limited co-promotion efforts in selected countries in the EU.

"This collaboration with Immatics gives us the opportunity to combine our unique technologies and expertise to create differentiated novel next-generation therapies. We very much look forward to this exciting partnership in the field of cancer immunotherapy," said Jan van de Winkel, Ph.D., Chief Executive Officer of Genmab.

Carsten Reinhardt, M.D., Ph.D., Chief Medical Officer and Managing Director of Immatics, commented: "We are very pleased to join forces with one of the world-leading biotechnology companies to develop and advance novel and highly active cancer therapeutics. This collaboration underpins Immatics’ leadership in intracellular tumor target identification and T-cell receptor engineering." Dr. Reinhardt further said: "Our bispecific TCR technology exhibits exceptional potency and favourable pharmacokinetic properties by combining Immatics’ proprietary T-cell engaging format with our high-affinity and highly specific T-cell receptors as reported at AACR (Free AACR Whitepaper) 20181."

Under the terms of the agreement, Genmab will pay Immatics an upfront fee of USD 54 million and Immatics is eligible to receive up to USD 550 million in development, regulatory and commercial milestone payments for each product, as well as tiered royalties on net sales.

Today’s news does not impact Genmab’s 2018 Financial Guidance.

New Preclinical Results on Onxeo’s AsiDNA™, First-in-Class DNA Repair Inhibitor, Point to Strong Synergy and Reversion of Tumor Resistance when combined to PARP inhibitors

On July 12, 2018 Onxeo S.A. (Euronext Paris, NASDAQ Copenhagen: ONXEO), ("Onxeo" or "the Company"), a clinical-stage biotechnology company specializing in the development of innovative drugs in oncology, in particular against rare or resistant cancers, reported new positive results from preclinical studies of AsiDNA, its first-in-class DNA Repair inhibitor, in combination with PARP inhibitors (PARPi) (Press release, Onxeo, JUL 12, 2018, View Source [SID1234527668]). The results of these extensive studies with different PARPi point to the ability of AsiDNA to prevent the occurrence of resistance and even to reverse the acquired resistance of the tumor cell after PARPi treatments. Furthermore, they show that the combination has a strong synergistic anti-tumor activity in in vitro and in vivo models of solid tumors resistant to PARPi (HR proficient). Together with the preliminary data on the activity and safety of AsiDNA expected in Q4 2018 from the DRIIV-1 clinical trial, these results support clinical development of AsiDNA in combination with PARP inhibitors, which should start from year-end 2018.

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!

Judith Greciet, Chief Executive Officer of Onxeo, said: "Onxeo is conducting an ambitious development program for AsiDNA, notably translational, in combination with various anti-cancer agents in order to provide strategic information aimed at determining the indications and combinations to target in further clinical development as soon as the first results from DRIIV-1 are available. Assessing the combination of AsiDNA with PARPi is a priority, as their mechanisms of action are very complementary in indications with high unmet medical needs. Sales for the PARPi class are already substantial in ovarian cancer and are expected to increase markedly in the near-term as products gain access to multiple additional oncology indications. Our recent studies indicate that AsiDNA in combination with PARPi could enable PARPi to overcome the requirement of a genetic mutation such as BRCA-, and show a strong synergistic activity versus PARPi alone. Moreover, the combination appears to both prevent the occurrence of resistance to PARPi and reverse the acquired resistance, which may considerably expand treatment duration with PARPi. A treatment combining AsiDNA and PARPi could therefore significantly broaden the patient population eligible to PARPi and improve their efficacy, which is of great interest to the scientific community, the pharmaceutical industry and the patients for its potential to address resistant cancers."

AsiDNA is a first-in-class DNA repair inhibitor in the field of DNA Damage Response (DDR) that mimics double-stranded DNA breaks in tumor cells, activating repair pathways, diverting repair enzymes from the target and finally depleting the cell through a unique mechanism of agonist and decoy.

Data show that in in vitro models of HR proficient TNBC (triple negative breast cancer) and SCLC (small cell lung cancer), AsiDNA maintains PARP1 expression, the repair enzyme inhibited by PARP inhibitors, and abrogates the occurrence of resistance to PARPi, including in models of cancers resistant to PARPi. Down regulation of the PARP1 enzyme is one of the mechanisms that supports the occurrence of resistance to PARPi inhibitors1. As AsiDNA hyper-activates repair enzymes, an up regulation of PARP1 expression following treatment with AsiDNA or with AsiDNA associated to PARPi support the use of AsiDNA to maintain the sensitivity to PARPi treatment.

Furthermore, combination treatment of olaparib with AsiDNA more than doubles the complete response rate observed with olaparib alone (71% vs. 33%) in an in vivo model of HR proficient TNBC model and inhibits tumor growth in a humanized Patient-Derived Xenograft (PDX) mice model of ovarian cancer resistant to olaparib. PDX models are considered highly predictive of clinical behavior2.

The Company will submit the detailed results of these preclinical studies to leading peer-reviewed publications and international scientific conferences.

Francoise Bono, Chief Scientific Officer of Onxeo, concluded: "These most recent data validate our disruptive approach to DNA-targeting and confirm the broad opportunities for our lead molecule thanks to its unique mechanism of action. Our team has built an extremely solid body of preclinical evidence, both in-vitro and in highly predictive humanized in-vivo models, which shows the potential of AsiDNA to reverse the resistance to PARP inhibitors and the strong synergy of their combination. This is the first part of our extensive translational plan which aims at confirming the full potential of AsiDNA in combination with other anticancer agents such as chemotherapies or epigenetic compounds, including belinostat. Additional data on these other possible combinations will be available after the summer to further inform the clinical development of AsiDNA in combinations offering the potential for significant therapeutic improvement."

Roche announces submission of supplemental New Drug Application for Venclexta for people with previously untreated acute myeloid leukaemia who are ineligible for intensive chemotherapy

On July 12, 2018 Roche (SIX: RO, ROG; OTCQX: RHHBY) reported submission of a supplemental New Drug Application (sNDA) to the United States (U.S.) Food and Drug Administration (FDA) for Venclexta (venetoclax), in combination with a hypomethylating agent or in combination with low dose cytarabine (LDAC), for treatment of people with previously untreated acute myeloid leukaemia (AML) who are ineligible for intensive chemotherapy (Press release, Hoffmann-La Roche, JUL 12, 2018, View Source [SID1234527663]). The submission is based on the results of two phase Ib/II studies that evaluated Venclexta in combination with azacitidine or decitabine (M14-358 study) or LDAC (M14-387 study) in this patient population. Venclexta is being developed by AbbVie and Roche. It is jointly commercialised by AbbVie and Genentech, a member of the Roche Group, in the U.S. and commercialised by AbbVie outside of the U.S.

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!

"Nearly 20,000 people will be diagnosed with AML in the U.S. this year, and many of them are not eligible to receive standard intensive chemotherapy," said Sandra Horning, MD, Roche’s Chief Medical Officer and Head of Global Product Development. "AML is an aggressive disease with the lowest survival rate of all leukaemias, and we look forward to working closely with the FDA to bring this potential option to patients with this very difficult-to-treat blood cancer as soon as possible."

Data recently presented from the phase Ib M14-358 study showed Venclexta in combination with azacitidine or decitabine resulted in a complete remission rate (with or without full recovery of normal blood cell count; CR/CRi) of 73% in patients treated with Venclexta at a dose of 400 mg.2 After more than a year of follow-up, the observed median overall survival (OS) across all Venclexta dose groups in the study was 17.5 months (95% CI: 12.3-not reached).2 The most common Grade 3-4 adverse events (occurring in 10% or more patients) were low white blood cell count with fever, low white blood cell count, anaemia, low platelet count and decreased potassium levels.2

Additionally, results from the phase Ib/II M14-387 study of Venclexta in combination with LDAC showed a CR/CRi rate of 62% in patients treated with Venclexta at a dose of 600 mg.3 After more than a year of follow-up, the observed median OS was 11.4 months (95% CI: 5.7-15.7).3 The most common Grade 3-4 adverse events (occurring in 10% or more patients) were low white blood cell count with fever, decreased potassium levels, pneumonia, disease progression, decreased phosphate levels, high blood pressure and sepsis (blood infection).3

The FDA previously granted two breakthrough therapy designations for Venclexta in previously untreated AML ineligible for intensive chemotherapy, either in combination with hypomethylating agents or LDAC, based on results from these two studies. Recently, the FDA approved Venclexta in combination with Rituxan (rituximab) for the treatment of people with chronic lymphocytic leukaemia (CLL) or small lymphocytic lymphoma (SLL), with or without 17p deletion, who have received at least one prior therapy. A robust clinical development programme is ongoing in several other cancer types.

About the M14-358 study
The M14-358 study (NCT02203773) is an open-label, phase Ib dose escalation and expansion study evaluating the safety and efficacy of Venclexta in combination with hypomethylating agents, azacitidine or decitabine, in 212 patients who are 60 years or older with previously untreated AML unfit to receive intensive chemotherapy. Study endpoints included CR/CRi, OS and safety.

About the M14-387 study
The M14-387 study (NCT02287233) is an open-label, phase Ib/II dose escalation and expansion study evaluating the safety and efficacy of Venclexta in combination with LDAC in 94 patients who are 60 years or older with previously untreated AML unfit to receive intensive chemotherapy. Study endpoints included CR/CRi, objective response rate (ORR), OS and safety.

About Venclexta
Venclexta is a small molecule designed to selectively bind and inhibit the BCL-2 protein, which plays an important role in a process called apoptosis (programmed cell death). Overexpression of the BCL-2 protein in AML has been associated with resistance to certain therapies. It is believed that blocking BCL-2 may restore the signalling system that tells cells, including cancer cells, to self-destruct. Venclexta is being developed by AbbVie and Roche. It is jointly commercialised by AbbVie and Genentech, a member of the Roche Group, in the U.S. and commercialised by AbbVie outside of the U.S.

Together, the companies are committed to further research with Venclexta, which is currently being evaluated in phase III clinical trials for several types of blood cancers. In the U.S., Venclexta has been granted four Breakthrough Therapy Designations by the FDA: in combination with Rituxan for people with relapsed or refractory CLL; as a monotherapy for people with relapsed or refractory CLL with 17p deletion; in combination with hypomethylating agents (azacitidine or decitabine) for people with untreated AML ineligible for intensive chemotherapy; and in combination with LDAC for people with untreated AML ineligible for intensive chemotherapy.

About Acute Myeloid Leukaemia
AML is an aggressive form of leukaemia that starts in immature forms of blood-forming cells, known as myeloid cells, found in the bone marrow.4 AML is the most common type of aggressive leukaemia in adults.1 It has the lowest survival rates of all types of leukaemia.5 Even with the best available therapies, older patients aged 65 and over have survival rates comparable to patients with advanced lung cancer, with a five year overall survival rate of <5%.6,7 Approximately 20,000 people in the U.S. and 18,000 in Europe are diagnosed with AML each year.8,9

NICE approves the targeted cancer immunotherapy, QARZIBA®? (dinutuximab beta) to treat children with high-risk neuroblastoma

On July 12, 2018 EUSA Pharma reported that a decision by the National Institute for Health and Care Excellence (NICE) to recommend the use of the targeted cancer immunotherapy, QARZIBA (dinutuximab beta) to treat children with high-risk neuroblastoma within the NHS in England and Wales (Press release, EUSA Pharma, JUL 12, 2018, View Source [SID1234527662]).[i] High-risk neuroblastoma is an aggressive form of neuroblastoma – the most common solid tumour of childhood that originates outside of the brain.[ii] Dinutuximab beta is the first targeted cancer immunotherapy approved for use on the NHS to treat this disease. It has been shown in a post-hoc analysis to improve overall survival (OS) outcomes compared to historically treated patients who did not receive immunotherapy as part of their care. Dinutuximab beta, when used in the maintenance phase of treatment for patients who did not receive prior immunotherapy, is also used to keep this cancer from returning or progressing in some children with high-risk neuroblastoma.ii

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!

"Today’s decision by NICE is a vital step forward in the treatment of young children with this aggressive type of cancer," said Dr Juliet Gray, Associate Professor in Paediatric Oncology at the Cancer Immunology Centre, University of Southampton. "By harnessing the body’s own immune system, dinutuximab beta has shown it can target and attack this cancer very effectively in some patients. For some children this could mean extra weeks or months with their families, for others it may even lead to them becoming cancer-free for a long period of time."

Dinutuximab beta is a monoclonal antibody (a type of protein) that binds to a specific target which is overexpressed on neuroblastoma cells, called GD2.[iii] This induces dual immune mechanisms that then enable the immune system to lead the destruction of neuroblastoma cancer cells.ii In the key phase III clinical study (APN311-302), a post hoc comparison of dinutuximab beta, used in the maintenance phase of the first-line treatment of high-risk neuroblastoma (n=367) , showed improved survival outcomes, with a 12% improvement in OS rate at three years versus using no immunotherapy in a historical control group of similar patients (n=450).ii The dinutuximab beta treated patients had an OS rate of around 65% at 5 years versus 50% compared to the historical control group (p=<0.0001).ii

Tony Heddon, Chair of Neuroblastoma UK commented: "Ensuring that children and families facing high-risk neuroblastoma have access to the medicines and care they need is absolutely critical. Today’s recommendation is a bold and forward-thinking decision from NICE and we applaud them, EUSA Pharma and all those across the community who have worked together to make this medicine available. This decision offers the hope that these children with high-risk neuroblastoma, may now have a better future in front of them."

On average, every week, two families in the UK will learn that their child has neuroblastoma, with approximately 100 children diagnosed each year.[iv] It is the most frequently-occurring solid tumour in infants under the age of one, accounting for around a fifth (22%) of all cancers diagnosed at this age.[v] Children with high-risk disease to whom this approval applies, account for approximately 40% of all neuroblastoma cases.[vi] Children with high-risk neuroblastoma typically undergo many rounds of complex and intensive treatment, usually comprising several cycles of chemotherapy, surgery, stem cell transplant and radiotherapy.[vii]

The recommendation from NICE within its Final Appraisal Determination (FAD) is that dinutuximab beta be used as an option for treating high-risk neuroblastoma after at least a partial response from induction chemotherapy, followed by myeloablative therapy and stem cell transplant in people aged 12 months and over, if the person has not had previous anti-GD2 immunotherapy.i

Lee Morley, CEO of EUSA Pharma added: "Today’s decision is the result of strong collaboration between NICE, EUSA Pharma and the neuroblastoma community, who have each worked tirelessly to ensure that every eligible child has the option to benefit from this potentially life-changing treatment. Our long-standing commitment has always been to secure access to dinutuximab beta for all eligible children with high-risk neuroblastoma, across the UK and today’s decision is a key part of that journey. Beyond England and Wales, we are continuing to work closely with the Scottish and Northern Irish health authorities with the aim of making this medicine available in those countries as quickly as possible."

This medicinal product is subject to additional monitoring. This will allow quick identification of new safety information. Adverse events should be reported. Reporting forms and information can be found at www.mhra.gov.uk/yellowcardreporting. Adverse events should also be reported to EUSA Pharma. Email: [email protected] Fax: +44(0)3305 001167

About dinutuximab beta

How it works

Dinutuximab beta is a monoclonal antibody (a type of protein) that has been designed to recognise and attach to a tumour-associated carbohydrate structure, called GD2, which is present in high amounts on the surface of neuroblastoma cells.ii When dinutuximab beta attaches to the neuroblastoma cells, it induces dual immune system mechanisms (the complement-dependant and antibody-dependant cell-mediated immune pathways) and makes them a target for the body’s immune system. This then mounts an attack to kill the cancer cells, using the body’s natural killer immune cells, and the complement protein system.ii

It’s development and approval

Dinutuximab beta is the result of a considered science–pharma collaboration. Dinutuximab beta was developed by Apeiron Biologics with a number of partners (in particular the SIOPEN academic neuroblastoma group) and acquired by EUSA Pharma in 2016, to bring the treatment to market. Dinutuximab beta received European approval in May 2017, first under the brand names dinutuximab beta Apeiron and Dinutuximab beta EUSA and subsequently under its new name, QARZIBA, approved by the European Medicines Agency in November 2017.iii

How it is taken

Dinutuximab beta is given as an infusion (drip) into a vein. Each course of treatment with the medicine is given for 5 or 10 days every 35 days. It is given for a total of 5 courses. The recommended dose depends on the patient’s weight and height.iii

Data supporting its use

Dinutuximab beta has been investigated in a number of clinical trials for high-risk neuroblastoma.ii During the NICE appraisal, the primary clinical evidence came from APN311-302, a multinational, open-label, randomised, controlled Phase III trial comparing dinutuximab beta plus isotretinoin (n=189) with dinutuximab beta plus isotretinoin plus interleukin-2 (n=190).i,ii The primary outcome in the trial was event-free survival at three years (disease progression or relapse, death and secondary tumour defined as events) with OS, overall response, and incidence of relapsed or refractory disease included as secondary outcomes.ii This study consisted of up to five different comparison phases, one of which was treatment with dinutuximab beta with or without interleukin-2 (IL-2) during the maintenance phase , in the first line setting.ii

In APN311-302, the 3-year event free survival (primary endpoint) showed rates of 55% without IL-2 and 61% with IL-2 (p=0.3202) while the 3-year OS rates were 64% and 69%, respectively (p=0.6114).i A comparison to an historical control group obtained from an earlier patient enrolment within the APN311-302 study (between 2002 and 2010) was performed using 450 high-risk neuroblastoma patients, who did not receive immunotherapy. Given the relatively high number of patients it is expected that these patients are representative of patients with high-risk neuroblastoma seen in clinical practice during this period. This comparison showed that the percentage of patients that were still alive after three years of follow-up was 12% higher after dinutuximab beta treatment (with or without IL-2) than for patients who did not receive immunotherapy, a difference considered clinically relevant.ii It also showed an OS rate of around 65% at 5 years with dinutuximab beta versus 50% in the historical control group (p=<0.0001).ii

At marketing authorisation, the European Medicines Agency considered that the available data set was not comprehensive and that measures were necessary to generate additional efficacy and safety data. EUSA Pharma is committed to this and is continuing to collect further data to widen the body of efficacy and safety information available on this medicine.ii

Side effects

Side effects with dinutuximab beta are common. In general, the most common side effects with dinutuximab beta (which may affect more than 7 in 10 people) are pyrexia (fever) and pain. Other side effects (which may affect more than 3 in 10 people) are hypersensitivity (allergy), vomiting, diarrhoea, capillary leak syndrome (leakage of fluid from blood vessels that can cause swelling and a drop in blood pressure) and hypotension (low blood pressure).iii

In the APN311-302 study, 98.9% of patients (362 of 366) in both treatment group experienced toxicities. Serious adverse events were reported more frequently in patients receiving IL-2 (46% of 183 patients) compared to patients not receiving IL-2 (27% of 183 patients). Serious adverse events leading to discontinuation of treatment were more frequent in the IL2 arm than the group without IL2,17% vs 6% of patients, respectively.ii