Cellectar Biosciences Announces Results From the First Two Cohorts of Its CLR 131 Phase 1 Trial: Demonstrates Excellent Efficacy, Overall Survival Benefit, and Progression Free Survival Similar to or Better than Recently Approved Therapies

On December 1, 2016 Cellectar Biosciences, Inc. (Nasdaq: CLRB) (the "company"), an oncology-focused, clinical stage biotechnology company, reported a data update on the first two cohorts of the company’s Phase I clinical study of CLR 131 in patients with relapsed or refractory multiple myeloma (Filing, 8-K, Cellectar Biosciences, DEC 1, 2016, View Source [SID1234516868]).

The clinical benefit rate for this study is 80 percent despite patients receiving an average of four prior treatments, including stem cell transplant and triple drug combinations. The patients in Cohort 1 received a single 12.5 mCi/m2 dose and patients in Cohort 2 received a single 18.75 mCi/m2 dose. At this time, Cohort 1 and Cohort 2 patients have demonstrated post treatment median survival of 11.9 months and 4.9 months, respectively. The median survival for all evaluable patients in both cohorts continues to increase and will be followed to determine overall survival benefit. Currently, the median overall survival (mOS) for each cohort is not yet evaluable. All evaluable patients in the clinical study experienced progression free survival (PFS). In Cohort 1, patients averaged 88.5 days of PFS. While patients in Cohort 2 have already achieved an average PFS of 127 days, the average PFS in Cohort 2 continues to increase as one of the four patients is still experiencing PFS. It is important to note that overall survival of 11.9 months and PFS of 127 days in this heavily pretreated patient population is better than or equivalent to that reported by several recently approved multiple myeloma drugs.

"The efficacy observed with CLR 131 at the 12.5 and 18.75 mCi/m2 single dose compares favorably to drugs recently approved for relapsed or refractory multiple myeloma. We believe that the 18.75 mCi/m2 dose could represent an acceptable single dose or multi-dose regimen for future studies," said Jim Caruso, president and CEO of Cellectar Biosciences. "Combined with its clean safety profile, we are optimistic regarding the potential of CLR 131 and look forward to seeing results from our recently initiated Cohort 3 at a single 25mCi/m2 dose."

An evaluation of adverse events between Cohort 1 and Cohort 2 reveal a similar profile. Patients in Cohort 1 experienced an average of 4.75 adverse events per patient while patients in Cohort 2 experienced an average of 4.25 events per patient. The median severity grade of the adverse events in both cohorts was 2.0 (mild to moderate), as graded by the National Cancer Institute’s Common Terminology Criteria for Adverse Events (NCI-CTCAE).

In the multi-center, open label Phase I dose escalation study, CLR 131 was administered as a single dose, 30-minute intravenous infusion on Day 1 with 40 mg dexamethasone orally weekly for 12 weeks. The primary study objective is to characterize the safety and tolerability of CLR 131 with and without dexamethasone in patients with relapsed and/or refractory multiple myeloma. Secondary study objectives include establishment of a recommended Phase II dose, both with and without dexamethasone, as well as an assessment of therapeutic activity, including progression-free survival (PFS) and additional efficacy endpoints.

Dose-escalation in this study uses a minimally modified, standard 3+3 schema with dose-limiting toxicities (DLTs) assessed through day 85 post-infusion. Each cohort consisted of four evaluable patients (three men, one woman in Cohort 1 and two men, two women in Cohort 2). Patients in both cohorts received an average of 4 prior treatments. All patients received and were relapsed or refractory to proteasome inhibitors and immunomodulatory drugs prior to enrollment, and all patients had received triple combination therapy as a single line of therapy at least once. One patient in Cohort 1 and three in Cohort 2 received autologous stem cell transplantation and three Cohort 1 patients and one Cohort 2 patient each received the latest approved drugs for multiple myeloma prior to enrollment. Patient’s ages range between 55-76 (Cohort 1) and 55-85 (Cohort 2) and averages were essentially identical at 68 and 69 years of age, respectively.

The company is currently enrolling patients into the study’s third cohort at a single 25 mCi/m2 dose and plans to provide an additional data update in the first half of 2017.

About CLR 131

CLR 131 is an investigational compound under development for a range of hematologic malignancies. It is currently being evaluated in a Phase I clinical trial in patients with relapsed or refractory multiple myeloma. The company plans to initiate a Phase II clinical study to assess efficacy in a range of B-cell malignancies in the first quarter of 2017. Based upon pre-clinical and interim Phase I study data, treatment with CLR 131 provides a novel approach to treating hematological diseases and may provide patients with therapeutic benefits, including overall response rate (ORR), an improvement in progression-free survival (PFS) and overall quality of life. CLR 131 utilizes the company’s patented PDC tumor targeting delivery platform to deliver a cytotoxic radioisotope, iodine-131 directly to tumor cells. The FDA has granted Cellectar an orphan drug designation for CLR 131 in the treatment of multiple myeloma.

About Phospholipid Drug Conjugates (PDCs)

Cellectar’s product candidates are built upon its patented cancer cell-targeting delivery and retention platform of optimized phospholipid ether-drug conjugates (PDCs). The company deliberately designed its phospholipid ether (PLE) carrier platform to be coupled with a variety of payloads to facilitate both therapeutic and diagnostic applications. The basis for selective tumor targeting of our PDC compounds lies in the differences between the plasma membranes of cancer cells compared to those of normal cells. Cancer cell membranes are highly enriched in lipid rafts, which are glycolipoprotein microdomains of the plasma membrane of cells that contain high concentrations of cholesterol and sphingolipids, and serve to organize cell surface and intracellular signaling molecules. PDCs have been tested in over 70 different xenograft models of cancer.

About Relapsed or Refractory Multiple Myeloma

Multiple myeloma is the second most common blood or hematologic cancer with approximately 30,000 new cases in the United States every year. It affects a specific type of blood cells known as plasma cells. Plasma cells are white blood cells that produce antibodies to help fight infections. While treatable for a time, multiple myeloma is incurable and almost all patients will relapse or the cancer will become resistant/refractory to current therapies.

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!


U.S. FDA Grants Fast Track Designation for HER2-Targeting Antibody Drug Conjugate DS-8201 for HER2-Positive Metastatic Breast Cancer

On December 1, 2016 Daiichi Sankyo Company, Limited (hereafter, Daiichi Sankyo) reported that the U.S. Food and Drug Administration (FDA) has granted Fast Track designation to its investigational HER2-targeting antibody drug conjugate DS-8201 for the treatment of HER2-positive unresectable and/or metastatic breast cancer in patients who have progressed after prior treatment with HER2-targeted therapies including ado-trastuzumab emtansine
(T-DM1) (Press release, Daiichi Sankyo, DEC 1, 2016, View Source [SID1234516875]).

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!



Fast Track designation is designed to facilitate the development and expedite the review of drugs that treat serious conditions and address an unmet medical need. The designation enables early and frequent communication with the FDA and is intended to accelerate drug approval and patient access to novel treatment options.

"This is an important milestone for DS-8201 that underscores the critical need to develop new and effective therapeutic options for patients with metastatic breast cancer whose tumors are no longer controlled by currently approved targeted HER2 treatments," said Antoine Yver, MD, MSc, Executive Vice President and Global Head, Oncology Research and Development, Daiichi Sankyo. "It’s our obligation to drive science forward to help bring innovative treatment options to cancer patients with the greatest unmet needs and we look forward to working closely with the FDA to optimize development of DS-8201."
Fast Track designation was granted based on results from the dose escalation part of a two-part phase 1 study that assessed the safety, tolerability and preliminary efficacy of DS-8201. These results were recently presented during a late-breaking poster discussion at the European Society for Medical Oncology (ESMO) (Free ESMO Whitepaper) 2016 Congress and further highlighted in the concluding summary session.1
The second part (dose expansion) of the ongoing phase 1 study is enrolling patients in Japan and the United States to evaluate the safety and efficacy of DS-8201 in four different cohorts of HER2 expressing cancers: patients with HER2+ metastatic breast cancer previously treated with T-DM1; patients with HER2+ gastric or gastroesophageal junction adenocarcinoma previously treated with trastuzumab; patients with HER2 low expressing breast cancer; and patients with other solid cancers that express HER2. For more information about the study visit ClinicalTrials.gov.

About DS-8201
DS-8201 is an investigational HER2-targeting antibody drug conjugate (ADC) currently in phase 1 clinical development for HER2+ advanced or metastatic breast cancer or gastric cancer, HER2 low expressing breast cancer and other HER2 expressing solid cancers.

DS-8201 is comprised of a humanized anti-HER2 antibody attached by a peptide linker to a novel topoisomerase I inhibitor that utilizes Daiichi Sankyo’s proprietary linker-payload technology, offering a unique mechanism of action.2 This linker-payload combination of DS-8201 allows for a higher drug-to-antibody ratio (DAR) of about 8, which may help target low expressing HER2 tumors by supplying more payload per antibody to a tumor.2

About HER2+ Metastatic Breast Cancer
Human epidermal growth factor receptor 2 (known as HER2) is a tyrosine kinase receptor growth-promoting protein found on the surface of some cancer cells.3 About one in five breast cancers overexpress the HER2/neu gene, which causes these cancers to grow more aggressively.3 Several unmet needs remain today in HER2+ metastatic breast cancer. Many tumors advance to the point where no currently approved HER2-targeted treatment continues to control the disease.4 Additionally, there are no existing options indicated for HER2 low expressing tumors (IHC2+/FISH- or IHC1+), which generally have poor prognosis.2,5

IBM and Pfizer to Accelerate Immuno-oncology Research with Watson for Drug Discovery

On December 1, 2016 IBM (NYSE: IBM) Watson Health and Pfizer Inc. (NYSE: PFE) reported a collaboration that will utilize IBM Watson for Drug Discovery to help accelerate Pfizer’s research in immuno-oncology, an approach to cancer treatment that uses the body’s immune system to help fight cancer (Press release, Pfizer, DEC 1, 2016, View Source [SID1234516872]). Pfizer is one of the first organizations worldwide to deploy Watson for Drug Discovery, and the first to customize the cloud-based cognitive tool – tapping in to Watson’s machine learning, natural language processing, and other cognitive reasoning technologies to support the identification of new drug targets, combination therapies for study, and patient selection strategies in immuno-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!



Immunotherapies, which modify a patient’s immune system to recognize and target cancer cells using a combination of vaccines, immunomodulators, and small/large molecules, are reshaping the field of oncology. Oncology researchers at Pfizer will use Watson for Drug Discovery to analyze massive volumes of disparate data sources, including licensed and publicly available data as well as Pfizer’s proprietary data. With this new tool, Pfizer researchers will analyze and test hypotheses to generate evidence-based insights for real-time interaction. The customized technology can also support efficient safety assessments.

Cancer is one of the leading causes of death worldwide, and is arguably one of the most complex diseases known to mankind.1 Many researchers believe that the future of immuno-oncology lies in combinations tailored to unique tumor characteristics, which could transform the cancer treatment paradigm and enable more oncology patients to be treated.

"Pfizer remains committed to staying at the forefront of immuno-oncology research," said Mikael Dolsten, President, Pfizer Worldwide Research & Development. "With the incredible volume of data and literature available in this complex field, we believe that tapping into advanced technologies can help our scientific experts more rapidly identify novel combinations of immune-modulating agents. We are hopeful that by leveraging Watson’s cognitive capabilities in our drug discovery efforts, we will be able to bring promising new immuno-oncology therapeutics to patients more quickly."

Laurie Olson, Executive Vice President, Strategy, Portfolio and Commercial Operations, Pfizer, said, "At Pfizer, we are entering a new frontier in data innovation in which we are investing in a range of new technologies and digital solutions to help us dynamically mine both internal and external data sources to find new connections in science, as well as help us better understand how diseases progress and how they could potentially be treated. Applying the power of cognitive computing to an area that is a core part of our DNA – discovering new medicines – is helping Pfizer to learn how we can most efficiently discover those immuno-oncology therapies that have the best chance of successful outcomes for patients."

The newly launched Watson for Drug Discovery is a cloud-based offering that aims to help life sciences researchers discover new drug targets and alternative drug indications. The average researcher reads between 200 and 300 articles in a given year2, while Watson for Drug Discovery has ingested 25 million Medline abstracts, more than 1 million full-text medical journal articles, 4 million patents and is regularly updated. Watson for Drug Discovery can be augmented with an organization’s private data such as lab reports and can help researchers look across disparate data sets to surface relationships and reveal hidden patterns through dynamic visualizations.

"We believe that the next great medical innovations will emerge as researchers and scientists find new patterns in existing bodies of knowledge. In order to do this, they need access to R&D tools that can help them efficiently navigate the opportunities and challenges presented by the explosion of data globally," said Lauren O’Donnell, Vice President of Life Sciences, IBM Watson Health. "IBM is honored to collaborate with Pfizer, and put Watson for Drug Discovery to work to support efforts in bringing life-saving immunotherapies to doctors and patients worldwide."

CTI BioPharma Presents Nonclinical Pharmacology Study Demonstrating Pacritinib Has Distinct Profile Amongst JAK Inhibitors At EORTC-NCI-AACR Molecular Targets And Cancer Therapeutics Symposium

On December 1, 2016 CTI BioPharma Corp. (CTI BioPharma) (NASDAQ and MTA: CTIC) reported the results of a translational pharmacology study comparing biomarker activity profiles for three JAK inhibitors: pacritinib, ruxolitinib and momelotinib, using the BioMAP Diversity PLUS panel of in vitro human primary cell-based systems (Press release, CTI BioPharma, DEC 1, 2016, View Source [SID1234516869]). The results demonstrated distinct profiles amongst these JAK inhibitors and suggest that clinical responses are likely to be distinct with each agent. The results were presented at the EORTC-NCI-AACR (Free EORTC-NCI-AACR Whitepaper) Molecular Targets and Cancer Therapeutics Symposium, November 29 – December 2 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!

At clinically relevant concentrations, each of the JAK inhibitors reduced inflammatory mediators associated with myelofibrosis such as TNF and IL-6, however they had divergent effects on other immunological and inflammatory pathways. When tested on human lymphoid cells, pacritinib had the strongest inhibitory activities on sIL-17A, sIL-2 and sIL-6, mediators involved in autoimmune responses, while ruxolitinib had the broader inhibitory activities in multiple systems. Both ruxolitinib and pacritinib were inhibitory to B cells, but only ruxolitinib inhibited T cells that are associated with cell-mediated immunity. Only pacritinib was anti-proliferative to endothelial cells and fibroblasts, effects commonly seen in agents with anti-cancer properties. The resulting distinct phenotypic profiles of pacritinib, ruxolitinib and momelotinib, illustrate that although all were developed as JAK2-ATP binding site inhibitors, they have divergent biological effects and likely will have distinct clinical activities.

The poster for Abstract #P094: "Comparative Biomarker Profiles of Pacrtitinib, Momelotinib, Pexidartinib and Ruxolitinib Using BIOMAP Diversity Plus Panel" is available at www.ctibiopharma.com.

About Pacritinib

Pacritinib is an investigational oral kinase inhibitor with specificity for JAK2, FLT3, IRAK1 and CSF1R. The JAK family of enzymes is a central component in signal transduction pathways, which are critical to normal blood cell growth and development, as well as inflammatory cytokine expression and immune responses. Mutations in these kinases have been shown to be directly related to the development of a variety of blood-related cancers, including myeloproliferative neoplasms, leukemia and lymphoma. In addition to myelofibrosis, the kinase profile of pacritinib suggests its potential therapeutic utility in conditions such as acute myeloid leukemia, or AML, myelodysplastic syndrome, or MDS, chronic myelomonocytic leukemia, or CMML, and chronic lymphocytic leukemia, or CLL, due to its inhibition of c-fms, IRAK1, JAK2 and FLT3.

Kura Oncology Presents Preclinical Data on KO-947 and Menin-MLL Inhibitor Program at the EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics

On December 1, 2016 Kura Oncology, Inc. (Nasdaq:KURA), a clinical stage biopharmaceutical company, reported preclinical data highlighting the identification and characterization of KO-947, its development candidate targeting ERK1/2 kinases (Press release, Kura Oncology, DEC 1, 2016, View Source [SID1234516867]). The company has also presented preclinical data relating to the identification and optimization of potent and selective inhibitors of the menin-MLL interaction. Both presentations took place at the EORTC-NCI-AACR (Free EORTC-NCI-AACR Whitepaper) Symposium on Molecular Targets and Cancer Therapeutics (EORTC) 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!

"We are excited to present preclinical data from these two innovative programs at EORTC, both of which showed compelling activity in preclinical models of cancer," said Yi Liu, Ph.D., Chief Scientific Officer. "Looking forward, we anticipate nominating a development candidate for our menin-MLL program by the end of 2016, and initiating a Phase 1 clinical trial for KO-947 in the first half of 2017."

KO-947 – A potent and selective inhibitor of ERK1/2 kinases

The RAS/RAF/MEK pathway is estimated to be activated in more than 30% of human cancers, including cancers arising from mutations in KRAS, NRAS and BRAF. Although inhibitors of both BRAF and MEK have been approved for treatment of melanoma, acquired resistance to these inhibitors has been documented both in preclinical and clinical samples due to reactivation of ERK1/2 kinases.

In preclinical studies presented today at EORTC, KO-947 showed potent inhibition of ERK signaling pathways and proliferation of tumor cells exhibiting dysregulation of MAPK pathway, including mutations in BRAF, NRAS or KRAS. KO-947 also inhibits MAPK signaling and cell proliferation in preclinical models that are resistant to BRAF and MEK inhibitors. Results obtained from screening a large panel of PDX models demonstrate that KO-947 induces tumor regressions in BRAF or RAS mutated tumor models as well as in tumor models lacking BRAF/RAS mutations but characterized by other dysregulation of the MAPK pathway.

KO-947 appears to be differentiated from other published ERK inhibitors by an extended residence time and prolonged pathway inhibition in vitro and in vivo. The data further suggest that the drug properties of KO-947 may allow Kura to maximize the therapeutic window with flexible administration routes and schedules, including intermittent dosing.

Inhibitors of the Menin-MLL Interaction

Chromosomal translocations that affect the mixed lineage leukemia (MLL) gene result in aggressive acute myeloid and lymphoid leukemias that are often resistant to standard chemotherapy. Approximately 5-10% of acute leukemias in adults, and 70% of acute leukemias in infants, are characterized by tumors with abnormal MLL fusions. MLL fusion proteins require menin for leukemogenic activity and selective disruption of the menin-MLL interaction represents a potential therapeutic approach for the treatment of acute leukemias with MLL rearrangements.

In preclinical studies presented at EORTC, inhibitors of the menin-MLL interaction showed potent inhibition of the proliferation of MLL leukemic cells. Inhibitors of the menin-MLL interaction displayed a greater than 50-fold reduction in potency in non-MLL-fusion leukemia cell lines and induced regression in a MV4:11 mouse xenograft model. The data show that the anti-tumor activity of menin-MLL inhibitors correlates with target engagement in tumors as well as inhibition of expression of downstream genes under the regulation of the fusion protein. Moreover, the inhibitors demonstrated potent efficacy in subcutaneous and disseminated models of MLL-fusion leukemias.

Both of the posters presented at EORTC can be found on Kura’s website in the Scientific Presentations and Papers section or by clicking here.