Virogin Biotech Ltd. Announces $5 Million in Series A+ Financing

On March 6, 2017 Virogin Biotech Ltd. ("Virogin") reported the completion of its Series A+ round of financing for a total of $5 million USD on February 28th, 2017 (Press release, Virogin Biotech, MAR 6, 2017, View Source [SID1234518865]). Virogin Biotech Ltd. specializes in cancertherapeutics research and discovery with a strong focus on innovative oncolyticvirotherapy.

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 financing was led by Sangel Capital and co-funded by bothexisting investors – Purity Star Ltd., Dahua Investment Corp. and Top FortuneVentures Ltd., and new strategic investor, FuRong Capital. FuRong Capital isthe industrial investment and commercialization arm of Shanghai FudanUniversity. The continued financial support from existing investors, alongsidethe endorsement by FuRong Capital, reflect investor confidence in Virogin’songoing development and further success.

The proceeds from this round offinancing will be used towards three areas: completion of pre-clinical research and IND enabling studies for VG161 – Virogin’s firstoncolytic virus product to enter clinical stage, continued expansion andoptimization of the research team with emphasis on clinical development andregulatory approval, and further development of the company’s product andproject pipelines.

"We are very grateful for the level of supportand dedication demonstrated by our existing investors during this round of financing,and we delightedly welcome FuRong Capital to the Virogin family." said Mr. Chris Huang, co-founder and CEO of Virogin, "Combinationtherapy will be the future direction of cancer immunotherapy, and oncolyticvirotherapy is inherently advantageous in achieving combination therapy;therefore, we expect it to be at the center of attention in the near future. Asone of the pioneers in the field of oncolytic virotherapy, Virogin possessesthe necessary technical expertise, proprietary technology and innovativeproduct design concept to be at the leading edge of this field. With thecapital infusion from this round of financing, we will be able to ensureprogress of our clinical and product development plans."

Dr. William Jia, co-founder and CSO of Virogin, also expressed hisexcitement, "We really appreciate the confidence and encouragement from theinvestors. In the past 20 months, Virogin has been growing at an incrediblepace, and has established its unique product pipeline with proprietarytechnology and innovative product design. Our first product VG161 showedexcellent pre-clinical results and we are looking forward to initiating clinicaltrials soon. The successful completion of this round of financing will fuel ourfast running engine to achieve higher speed and reach new heights. "

Dr. NanxingHe, Managing Partner at Sangel Capital, added, "As one of the earliestinvestors of Virogin, we have witnessed the level of progress the companyachieved in less than 2 years, and we sincerely admire their dedication and entrepreneurship.Dr. Jia and his research team are both knowledgeable and trustworthy. As aprofessional investment institution in the field of biomedicine, we willcontinue to be a strong supporter of Virogin, and we look forward to a brightfuture for the company."

"Weare delighted to participate in Virogin’s Series A+ round of financing," statedDr. Pengjun Sun, General Manager of FuRong Capital, "Better yet, Virogin isco-founded by a distinguished scientist and a seasoned entrepreneur, both ofwhom are alumni of Shanghai Fudan University. They have demonstrated strongexpertise and experience in their respective fields, thus we are confident inVirogin’s market competitiveness and growth potential in the biotech andpharmaceutical industry. We look forward to further collaboration betweenVirogin and Shanghai Fudan University in the areas of research and discovery,human capital, and clinical development. Collectively, we will be able tofurther promote the growth and development of Virogin."

Deciphera Pharmaceuticals Initiates a Phase 1 Clinical Trial of DCC-3014

On March 6, 2017 Deciphera Pharmaceuticals, a clinical-stage biopharmaceutical company focused on addressing key mechanisms of tumor drug resistance, reported that it has initiated a multi-center Phase 1 clinical trial of DCC-3014, a highly-selective small molecule CSF1R inhibitor, in patients with advanced malignancies (Press release, Deciphera Pharmaceuticals, MAR 6, 2017, View Source [SID1234518029]).

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 initiate this first-in-human Phase 1 clinical trial of DCC-3014, our small molecule switch control inhibitor of CSF1R," said Michael D. Taylor, Ph.D., Deciphera’s President and Chief Executive Officer. "Preclinical data from a number of cancer models have demonstrated that DCC-3014 has potent macrophage checkpoint inhibitory activity. We believe DCC-3014 has great potential as a novel immunomodulatory agent and an important new therapy for cancer patients."

The Phase 1 clinical trial is designed to evaluate the safety, pharmacokinetics and pharmacodynamics of multiple ascending doses of DCC-3014. Up to 55 patients with advanced malignancies will be enrolled across both dose-escalation and expansion phases of the study. For more information about the clinical trial design please visit www.clinicaltrials.gov.

"DCC-3014 inhibits tumor associated macrophages which are believed to play a key role in the tumor microenvironment," said Oliver Rosen, M.D., Chief Medical Officer at Deciphera. "By blocking macrophage immune checkpoints, DCC-3014 has demonstrated additive or synergistic immunomodulatory activity in combination with PD-1 inhibition in multiple preclinical models."

About DCC-3014
DCC-3014 was purposefully designed using the company’s proprietary Switch Control Inhibitor platform to be a highly-specific macrophage immunomodulatory agent. In preclinical models, DCC-3014 has demonstrated potent inhibition of the colony stimulating factor 1 receptor (CSF1R), an important target for the treatment of many cancer indications. Deciphera reported preclinical data demonstrating potent macrophage checkpoint inhibition with DCC-3014 in multiple cancer models both as a single agent and in combination with a PD1 inhibitor at the American Association for Cancer Research (AACR) (Free AACR Whitepaper) Annual Meeting in April 2016. DCC-3014 is currently in Phase 1 clinical development in patients with advanced malignancies.

MAST THERAPEUTICS REPORTS FOURTH QUARTER AND FULL YEAR 2016 FINANCIAL RESULTS

On March 6, 2017 Mast Therapeutics, Inc. (NYSE MKT: MSTX), a clinical-stage biopharmaceutical company, reported financial results for the fourth quarter and year ended December 31, 2016 (Filing, Q4/Annual, Mast Therapeutics, 2016, MAR 6, 2017, View Source [SID1234518024]).

“We are pleased with the progress made with AIR001 during the year and anticipate the closing of the merger with Savara to take place in the second quarter of this year,” stated Brian M. Culley, Chief Executive Officer. “We believe this merger offers our stockholders a diversified, late-stage product development pipeline with important forthcoming milestones.”

Fourth Quarter 2016 Operating Results

The Company’s net loss for the fourth quarter of 2016 was $6.0 million, or $0.02 per share (basic and diluted), compared to a net loss of $10.2 million, or $0.06 per share (basic and diluted), for the same period in 2015.

The Company recognized $83,000 of revenue for the fourth quarter of 2016, representing reimbursement of costs related to the nonclinical study of vepoloxamer that is being funded by a Small Business Innovation Research (SBIR) grant. The Company recognized no revenue for the same period in 2015.

Research and development (R&D) expenses for the fourth quarter of 2016 were $78,000, a decrease of approximately $7.1 million, or 99%, compared to $7.2 million for the same period in 2015. This reduction in R&D expense was due principally to the Company’s decision to discontinue clinical development of vepoloxamer in September 2016. External clinical study fees and expenses decreased by $3.9 million, external nonclinical study fees and expenses decreased by $2.9 million and R&D personnel costs decreased by $0.3 million for the fourth quarter of 2016 compared to the same period in 2015.

Selling, general and administrative (SG&A) expenses for the fourth quarter of 2016 were $1.9 million, a decrease of $0.6 million, or 23%, compared to $2.5 million for the same period in 2015. The decrease was primarily due to reduced fees for consulting and legal services and personnel costs compared to the 2015 period.

Interest expense was $0.2 million for the fourth quarter of 2016, compared to $0.5 million for the same period in 2015. The decrease in interest expense was primarily due to the Company’s prepayment of $10.0 million of the principal balance of its debt facility in October 2016, which lowered the overall principal balance of the debt significantly. The principal balance was $3.3 million at December 31, 2016 compared to $15.0 million at December 31, 2015.

Fiscal Year 2016 Financial Results

The Company’s net loss for the year ended December 31, 2016 was $36.1 million, or $0.17 per share (basic and diluted), compared to a net loss of $39.8 million, or $0.25 per share (basic and diluted), for the same period in 2015.

The Company recognized $128,000 of revenue for the year ended December 31, 2016, representing reimbursement of costs related to the nonclinical study of vepoloxamer that is being funded by a SBIR grant. The Company recognized no revenue for the same period in 2015.

R&D expenses for the year ended December 31, 2016 were $20.8 million, a decrease of $7.5 million, or 26%, compared to $28.3 million for the same period in 2015. The decrease was due primarily to a $4.1 million decrease in external nonclinical study fees and expenses, a $3.3 million decrease in external clinical study fees and expenses, and a $0.3 million decrease in personnel costs, offset by a $0.3 million increase in share-based compensation expense.

The decrease in external nonclinical study fees and expenses resulted primarily from decreases in research-related manufacturing costs for vepoloxamer ($4.7 million) and nonclinical studies of vepoloxamer ($1.5 million), offset by

increased costs related to preparing a new drug application for vepoloxamer ($1.9 million), which project was discontinued in September 2016, and research-related manufacturing costs for AIR001 ($0.2 million). The decrease in external clinical study fees and expenses was due primarily to decreases in costs for the Phase 3 study of vepoloxamer in sickle cell disease ($5.0 million) and the Phase 2 study of vepoloxamer in ALI that was discontinued in the third quarter of 2015 ($0.5 million), offset by increased costs related to the Phase 2 study of vepoloxamer in heart failure ($1.3 million) and the investigator-sponsored Phase 2 studies of AIR001 in HFpEF ($0.9 million). The $0.3 million decrease in personnel costs was due primarily to reductions in the Company’s workforce that occurred in the fourth quarter of 2016.

SG&A expenses for the year ended December 31, 2016 were $9.3 million, a decrease of $1.7 million, or 15%, compared to $11.0 million for the same period in 2015. This decrease was due primarily to a $1.0 million decrease in personnel costs and a $0.5 million decrease in professional and consulting fees.

Interest expense was $2.1 million for the year ended December 31, 2016, an increase of $1.5 million compared to $0.6 million for the same period in 2015. The increase in interest expense was primarily due to interest expense on a $15 million principal balance under the Company’s debt facility for nine months in 2016 versus approximately four months of interest expense on the debt facility in 2015, as well as increased amortization of debt issuance costs as a result of a change in the amortization schedule of such costs due to prepayment of $10 million of the principal balance in October 2016.

FDA Lifts Clinical Hold on Seattle Genetics’ Phase 1 Trials of Vadastuximab Talirine

On March 6, 2017 Seattle Genetics, Inc. (Nasdaq: SGEN), a global biotechnology company, reported that the U.S. Food and Drug Administration (FDA) has lifted the clinical hold announced on December 27, 2016 on phase 1 trials of vadastuximab talirine (SGN-CD33A; 33A) in acute myeloid leukemia (AML) (Press release, Seattle Genetics, MAR 6, 2017, View Source [SID1234518019]).

“The clinical hold on our early-stage vadastuximab talirine clinical trials has been resolved through a comprehensive analysis of the clinical data from over 300 patients treated to date, evaluation by an independent committee of clinical experts, collaborative interactions with the FDA, and protocol amendments designed to further enhance patient safety,” said Jonathan Drachman, M.D., Chief Medical Officer and Executive Vice President, Research and Development at Seattle Genetics. “We will resume two phase 1 trials in AML and plan to initiate a randomized phase 2 trial during 2017 evaluating vadastuximab talirine in combination with standard of care chemotherapy in frontline, younger AML patients. In addition, we are continuing to enroll our ongoing phase 3 randomized CASCADE trial in frontline older AML patients and our phase 1/2 trial in frontline high-risk myelodysplastic syndrome (MDS).”

Seattle Genetics will resume two phase 1 trials of vadastuximab talirine. The first is combination treatment with standard of care, or 7+3, chemotherapy in newly diagnosed younger AML patients and the second is monotherapy and combination treatment with hypomethylating agents in both newly diagnosed and relapsed AML patients. Seattle Genetics will not resume the phase 1/2 trial of vadastuximab talirine monotherapy in pre- and post-allogeneic transplant AML patients given the challenges of developing therapies in this specific setting. The company’s randomized global phase 3 CASCADE trial in frontline older AML and phase 1/2 trial in frontline MDS were not placed on clinical hold and have continued to enroll patients. Planned studies include a randomized phase 2 trial of vadastuximab talirine in combination with 7+3 chemotherapy in frontline younger AML patients. Going forward, additional risk mitigation measures will be implemented in all vadastuximab talirine studies, including revised eligibility criteria and stopping rules for veno-occlusive disease (VOD).

About Vadastuximab Talirine
Vadastuximab talirine (SGN-CD33A; 33A) is a novel investigational ADC targeted to CD33 utilizing Seattle Genetics’ proprietary ADC technology. CD33 is expressed on most AML and MDS blast cells. The CD33 engineered cysteine antibody is stably linked to a highly potent DNA binding agent called a pyrrolobenzodiazepine (PBD) dimer via site-specific conjugation technology (EC-mAb). PBD dimers are significantly more potent than systemic chemotherapeutic drugs and the EC-mAb technology allows uniform drug-loading onto an ADC. The ADC is designed to be stable in the bloodstream and to release its potent cell-killing PBD agent upon internalization into CD33-expressing cells.
Vadastuximab talirine was granted Orphan Drug Designation by both the U.S. Food and Drug Administration (FDA) and the European Commission for the treatment of AML. FDA orphan drug designation is intended to encourage companies to develop therapies for the treatment of diseases that affect fewer than 200,000 individuals in the United States.

Pieris Pharmaceuticals Announces Presentation of IND-enabling Data for Bispecific Immuno-Oncology Drug Candidate, PRS-343, at the 2017 Meeting of the American Association for Cancer Research (AACR)

On March 6, 2017 Pieris Pharmaceuticals, Inc. (NASDAQ: PIRS), a clinical-stage biotechnology company advancing novel biotherapeutics through its proprietary Anticalin technology platform, reported that a set of ex vivo and in vivo data informing the trial design of a first-in-patient clinical trial for PRS-343, a first-in-class 4-1BB/HER2 bispecific, will be presented in a poster session at the 2017 Annual Meeting of the American Association for Cancer Research (AACR) (Free AACR Whitepaper) to be held in Washington D.C., April 1 – 5, 2017 (Press release, Pieris Pharmaceuticals, MAR 6, 2017, View Source [SID1234518017]). PRS-343 binds to 4-1BB (CD137) on T cells and HER2 on tumor cells, producing tumor-targeted immune activation.

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!

Further details include:

Poster Number : 16

Title : Preclinical toxicology and pharmacology for the 4-1BB/HER2 bispecific PRS-343: A first-in-class costimulatory T cell engager.

Date and Time : Tuesday April 4, 2017, 8:00 AM – 12:00 PM

Location : Walter E. Washington Convention Center, Halls A-C, Poster Section 27
About PRS-343:
PRS-343 is a bispecific monoclonal antibody/Anticalin fusion protein comprised of a HER2 tumor-targeting mAb genetically linked to a potent Anticalin specific for the immune costimulatory TNF family receptor 4-1BB (CD137). PRS-343 is being developed as the first 4-1BB based therapeutic to mediate the activation of tumor-specific T lymphocytes selectively within the tumor microenvironment (TME). 4-1BB is a potent costimulatory immunoreceptor and an established marker for tumor-specific infiltrating T lymphocytes (TILs), and is, therefore, an attractive target for cancer immunotherapy. In in vivo preclinical tumor models, PRS-343 has demonstrated potent T lymphocyte activation localized to the TME of established HER2-positive tumors, indicating the potential for both enhanced safety and efficacy.