Feeds:
Posts
Comments

Posts Tagged ‘cancer vaccine’

Real Time Coverage @BIOConvention #BIO2019: International Cancer Clusters Showcase June 3, Philadelphia PA

Reporter: Stephen J. Williams PhD @StephenJWillia2

Updated on 07/08/2021

https://cancerdiscovery.aacrjournals.org/content/early/2021/07/01/2159-8290.CD-20-1741

Updated on 07/08/2021

https://medicalxpress.com/news/2021-07-cancer-wider-access-immunotherapy.html

Larry Blandford PharmD from Precision Medicine Group gave introduction about development of precision oncology medicine.  Talked about value and value determination for partnerships.

Company Pitches:

Kernal Biologics: Preclinical immunotherapy company developing mRNA therapeutics.  Their therapy only have activity in p53 deficient cells (messenger 2.0).  They identified, by screening, multiple mRNAs that have oncoselectivity; ONC-333 is their lead mRNA active in AML and NSCLC.  Looking for 5.5M seed $

Vaccibody AS: Vaccine technology from Oslo University to target antigen to antigen presenting cells.  They are targeting the myocytes and dimerize the antigen to MHC.  Targeting melanoma, certain cervical cancers, and hemotologic cancers.  Technology based on identified neoantigens obtained from tumor biopsy.Three vaccines: VB10.neo  VB10.16 against HPV cervical

Chimeric Therapeutics: developing CART to solid malignancies against CLEC14 (tumor endothelial marker), may make tumor susceptible to hypoxia.  Targeting pancreatic cancer, prelim results in mice , efficacy of 15%, working on 3rd generation CART

Memo Therapeutics: Antibody therapeutics; based on Dropzylla single B cell sorting and subsequent screening for mAb.  Targeting checkpoint inhibitors on solid tumors;  have a new target other than PD1; target undisclosed on NK cells and T cells; Early stage have academic partners; seeking 20Million Swiss Francs

Takeda Oncology: Chris Hurff Senior Director Business Development; they depend on partnerships as they feel internal RD is less effective.  They are diversifying their portfolio from small molecules. They have over 200 partnerships (132 in Boston). They are focusing on heme, lung, and Immunooncology. Partnering model: CEI (center external innovation) deals with both academic and small biotechs.  They have numerous partners including Shatto and MD Anderson.

Read Full Post »

Immunoediting can be a constant defense in the cancer landscape

Immuno-editing can be a constant defense in the cancer landscape, Volume 2 (Volume Two: Latest in Genomics Methodologies for Therapeutics: Gene Editing, NGS and BioInformatics, Simulations and the Genome Ontology), Part 1: Next Generation Sequencing (NGS)

Reporter and Curator: Dr. Sudipta Saha, Ph.D.

 

There are many considerations in the cancer immunoediting landscape of defense and regulation in the cancer hallmark biology. The cancer hallmark biology in concert with key controls of the HLA compatibility affinity mechanisms are pivotal in architecting a unique patient-centric therapeutic application. Selection of random immune products including neoantigens, antigens, antibodies and other vital immune elements creates a high level of uncertainty and risk of undesirable immune reactions. Immunoediting is a constant process. The human innate and adaptive forces can either trigger favorable or unfavorable immunoediting features. Cancer is a multi-disease entity. There are multi-factorial initiators in a certain disease process. Namely, environmental exposures, viral and / or microbiome exposure disequilibrium, direct harm to DNA, poor immune adaptability, inherent risk and an individual’s own vibration rhythm in life.

 

When a human single cell is crippled (Deranged DNA) with mixed up molecular behavior that is the initiator of the problem. A once normal cell now transitioned into full threatening molecular time bomb. In the modeling and creation of a tumor it all begins with the singular molecular crisis and crippling of a normal human cell. At this point it is either chop suey (mixed bit responses) or a productive defensive and regulation response and posture of the immune system. Mixed bits of normal DNA, cancer-laden DNA, circulating tumor DNA, circulating normal cells, circulating tumor cells, circulating immune defense cells, circulating immune inflammatory cells forming a moiety of normal and a moiety of mess. The challenge is to scavenge the mess and amplify the normal.

 

Immunoediting is a primary push-button feature that is definitely required to be hit when it comes to initiating immune defenses against cancer and an adaptation in favor of regression. As mentioned before that the tumor microenvironment is a “mixed bit” moiety, which includes elements of the immune system that can defend against circulating cancer cells and tumor growth. Personalized (Precision-Based) cancer vaccines must become the primary form of treatment in this case. Current treatment regimens in conventional therapy destroy immune defenses and regulation and create more serious complications observed in tumor progression, metastasis and survival. Commonly resistance to chemotherapeutic agents is observed. These personalized treatments will be developed in concert with cancer hallmark analytics and immunocentrics affinity and selection mapping. This mapping will demonstrate molecular pathway interface and HLA compatibility and adaptation with patientcentricity.

References:

 

https://www.linkedin.com/pulse/immunoediting-cancer-landscape-john-catanzaro/

 

https://www.cell.com/cell/fulltext/S0092-8674(16)31609-9

 

https://www.researchgate.net/publication/309432057_Circulating_tumor_cell_clusters_What_we_know_and_what_we_expect_Review

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4190561/

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5840207/

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5593672/

 

https://www.frontiersin.org/articles/10.3389/fimmu.2018.00414/full

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5593672/

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4190561/

 

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4388310/

 

https://www.linkedin.com/pulse/cancer-hallmark-analytics-omics-data-pathway-studio-review-catanzaro/

 

Read Full Post »

Moderna Therapeutics Deal with Merck: Are Personalized Vaccines here?

Curator & Reporter: Stephen J. Williams, Ph.D.

UPDATED:  3/11/2023

Source: https://www.fiercebiotech.com/biotech/chasing-moderna-merck-pays-50m-join-race-develop-cancer-preventing-vaccine

Chasing Moderna, Merck pays $50M to join race to develop cancer-preventing vaccine

Merck & Co.
Merck’s deal with ModeX Therapeutics is a win for Opko Health, which bought ModeX for $300 million in stock last year. (Photo by Kena Betancur/Getty Images)

Merck & Co. has joined the emerging race to develop an Epstein-Barr virus (EBV) vaccine, paying ModeX Therapeutics $50 million upfront and dangling $872.5 million in biobucks for global rights to a preclinical challenger to shots in clinical development at Moderna and the National Institutes of Health (NIH).

EBV infects most people at some point in their lives. Many people have no symptoms, but the virus can cause infectious mononucleosis, a condition associated with fatigue and fever as well as linked to a range of other diseases. Carried as an asymptomatic infection, the virus is associated with 200,000 cancer cases a year, and a paper published last year showed it greatly increases the risk of multiple sclerosis.

Merck, which pioneered the idea of vaccinating to prevent cancer with HPV shot Gardasil, sees the evidence as an opportunity—and has identified ModeX as the company to help it realize that opportunity. The deal with ModeX gives Merck an exclusive worldwide license to the preclinical vaccine candidate MDX-2201.

ModeX developed MDX-2201 using a modular nanoparticle vaccine platform. The approach has resulted in a vaccine that presents antigens from four viral proteins—gH, gL, gp42 and gp350—involved in viral entry into host cells. By targeting four proteins, the vaccine could inhibit the infection of both B cells and epithelial cells.

The vaccine is differentiated from other candidates. Moderna’s mRNA-1189 uses lipid nanoparticles to get mRNA encoding for four viral proteins—gp42, gp220, gH and gL—into human cells. Other candidates, including the NIH’s clinical-phase prospect, target gp350, the most abundant glycoprotein on the EBV envelope, although this approach has so far failed to yield an effective vaccine.

Merck sees potential in ModeX’s broader approach and will work with the biotech to get the prospect ready for the clinic. Tarit Mukhopadhyay, Ph.D., vice president of infectious diseases and vaccine discovery at Merck Research Laboratories, highlighted the company’s experience with Gardasil in a statement to disclose the deal.

“We have a proud legacy of developing vaccines including several that have the potential to help protect against certain types of cancer. We look forward to working with the ModeX Therapeutics team to apply our experience and expertise to evaluate the potential of MDX-2201 to help protect against EBV infection and other, potentially related, conditions,” Mukhopadhyay said.

The deal is a win for Opko Health, which bought ModeX for $300 million in stock last year. In addition to MDX-2201, the takeover gave Opko control of a tetra-specific antibody for treating solid tumors that is on course to enter the clinic next year.

Take aways:

  • RNA based vaccines are a cost-effective method of developing and manufacturing a personalized cancer vaccine strategy; traditional vaccine methodology has not been met with much success as a cancer therapeutic
  • Most of the older RNA vaccine technology depended on isolated dendritic cells or T cell populations and ex-vivo treatment with RNA vaccine, HOWEVER, Moderna has developed a technology that circumvents the need for ex-vivo vaccination
  • There are multiple companies involved in this new RNA strategy (Moderna, Caperna {now Moderna}, CureVac, Biontech)

From BusinessWire at http://www.businesswire.com/news/home/20160629005446/en/Merck-Moderna-Announce-Strategic-Collaboration-Advance-mRNA-Based

Merck and Moderna Announce Strategic Collaboration to Advance Novel mRNA-Based Personalized Cancer Vaccines with KEYTRUDA®(pembrolizumab) for the Treatment of Multiple Types of Cancer

Collaboration Combines Merck’s Leadership in Immuno-Oncology with Moderna’s Pioneering mRNA Vaccine Technology and Rapid Cycle Time, Small-Batch GMP Manufacturing Capabilities

KENILWORTH, N.J. & CAMBRIDGE, Mass.–(BUSINESS WIRE)–Merck (NYSE:MRK), known as MSD outside the United States and Canada, and Moderna Therapeutics today announced a strategic collaboration and license agreement to develop and commercialize novel messenger RNA (mRNA)-based personalized cancer vaccines. The collaboration will combine Merck’s established leadership in immuno-oncology with Moderna’s pioneering mRNA vaccine technology and GMP manufacturing capabilities to advance individually tailored cancer vaccines for patients across a spectrum of cancers.

“Combining immunotherapy with vaccine technology may be a new path toward improving outcomes for patients”

Tweet this

Moderna and Merck will develop personalized cancer vaccines that utilize Moderna’s mRNA vaccine technology to encode a patient’s specific neoantigens, unique mutations present in that specific patient’s tumor. When injected into a patient, the vaccine will be designed to elicit a specific immune response that will recognize and destroy cancer cells. The companies believe that the mRNA-based personalized cancer vaccines’ ability to specifically activate an individual patient’s immune system has the potential to be synergistic with checkpoint inhibitor therapies, including Merck’s anti-PD-1 therapy, KEYTRUDA® (pembrolizumab). In addition, Moderna has developed a rapid cycle time, small-batch manufacturing technique that will uniquely allow the company to supply vaccines tailored to individual patients within weeks.

Under the terms of the agreement, Merck will make an upfront cash payment to Moderna of $200 million, which Moderna will use to lead all research and development efforts through proof of concept. The development program will entail multiple studies in several types of cancer and include the evaluation of mRNA-based personalized cancer vaccines in combination with Merck’s KEYTRUDA® (pembrolizumab). Moderna will also utilize the upfront payment to fund a portion of the build-out of a GMP manufacturing facility in suburban Boston for the purpose of personalized cancer vaccine manufacturing.

Following human proof of concept studies, Merck has the right to elect to make an additional undisclosed payment to Moderna. If exercised, the two companies will then equally share cost and profits under a worldwide collaboration for the development of personalized cancer vaccines. Moderna will have the right to elect to co-promote the personalized cancer vaccines in the U.S. The agreement entails exclusivity around combinations with KEYTRUDA. Moderna and Merck will each have the ability to combine mRNA-based personalized cancer vaccines with other (non-PD-1) agents.

Combining immunotherapy with vaccine technology may be a new path toward improving outcomes for patients,” said Dr. Roger Perlmutter, President, Merck Research Laboratories. “While the area of personalized cancer vaccine research has faced challenges in the past, there have been many recent advances, and we believe that working with Moderna to combine an immuno-oncology approach, using KEYTRUDA, with mRNA-based personalized cancer vaccines may have the potential to transform the treatment of cancer.”

“Our team has made significant progress since beginning our work in personalized cancer vaccines just last year. Through this collaboration with Merck, we are now well-positioned to accelerate research and development with a goal of entering the clinic in 2017, as well as to apply our unique GMP manufacturing capabilities to support the rapid production of these highly individualized vaccines,” said Stéphane Bancel, chief executive officer of Moderna. “We value our continued collaboration with Merck, and we look forward to working together to harness the potential of personalized cancer vaccines and immuno-oncology to bring a new treatment paradigm to patients.”

Merck and Moderna have an existing collaboration and license agreement focused on the discovery and development of mRNA-based infectious disease vaccines and passive immunity treatments. Moderna is also advancing its own pipeline of infectious disease vaccine candidates and currently has two phase 1 studies underway in Europe and the U.S.

About Moderna Therapeutics

Moderna is a clinical stage pioneer of messenger RNA Therapeutics™, an entirely new in vivo drug technology that produces human proteins, antibodies and entirely novel protein constructs inside patient cells, which are in turn secreted or active intracellularly. This breakthrough platform addresses currently undruggable targets and offers a potentially superior alternative to existing drug modalities for a wide range of diseases and conditions. Moderna is developing and plans to commercialize its innovative mRNA drugs through its own ventures and its strategic relationships with established pharmaceutical and biotech companies. Its current ventures are:

  • Onkaido, focused on oncology,
  • Valera, focused on infectious diseases,
  • Elpidera, focused on rare diseases, and
  • Caperna, focused on personalized cancer vaccines.

Cambridge-based Moderna is privately held and currently has strategic agreements with AstraZeneca, Alexion Pharmaceuticals and Merck. To learn more, visit www.modernatx.com.

From the Moderna Therapeutics Website

Our mRNA Platform

At Moderna, we are pioneering the development of a new class of drugs made of messenger RNA (mRNA). This novel drug platform builds on the discovery that modified mRNA can direct the body’s cellular machinery to produce nearly any protein of interest, from native proteins to antibodies and other entirely novel protein constructs that can have therapeutic activity inside and outside of cells.

Our efforts are helping Moderna and the industry to flatten the mRNA learning curve across the full breadth of competencies needed to drive the platform forward, including chemistry, mRNA biology, formulation, process development, automation and high-throughput production, quality, and Good Manufacturing Practice (GMP) manufacturing.

Drug Modalities

Building from our mRNA core expression platform, we have created a new scale of drug discovery and development that enables a series of new drug modalities. Each modality represents a distinct approach to using the mRNA platform to encode proteins that achieve a therapeutic benefit, enabling us to develop numerous drug candidates across a wide array of therapeutic areas.

Vaccines

Vaccines are substances that teach the immune system to rapidly recognize and destroy invading pathogens such as bacteria or viruses, preparing the body’s adaptive immunity for future exposure to the pathogen. Historically, vaccines have introduced immune-activating markers from pathogens into the body. Conversely, Moderna is developing mRNA-based vaccines that enable the body to produce and present immunogenic proteins to the immune system.

Moderna is also developing mRNA-based personalized cancer vaccines to prime the immune system to recognize cancer cells and mount a strong, tailored response to each individual patient’s cancer. Moderna’s technology allows for a rapid turn-around time in production of these unique mRNA vaccines.

Intracellular/Transmembrane

Many diseases are caused by defects in proteins that function inside cells. Existing methods of protein-based therapy do not allow for proteins to reach the intracellular space, and as such are unable to replace the defective, disease-causing proteins within cells. Moderna’s platform allows for the development of mRNA therapies that can stimulate production of intracellular proteins as well as transmembrane proteins. This could potentially lead to a novel approach to treating a vast array of rare genetic and other diseases caused by intracellular protein defects.

Intratumoral

Many targets for the treatment of cancer have been identified but their therapeutic potential has been limited by either the inability to access these targets, or by systemic toxicities. Moderna’s platform allows for localized expression of therapeutic proteins within the tumor microenvironment.

Secreted antibodies

Antibodies are secreted proteins that bind to and inhibit specific targets. Moderna’s platform has the potential to stimulate the body’s own cells to produce specific antibodies that can bind to cellular targets.

Secreted proteins

Proteins are large, complex molecules that have many critical functions both inside and outside of cells. Moderna’s platform stimulates cells to produce and secrete proteins that can have a therapeutic benefit through systemic exposure.

Moderna is comprised of four smaller companies, the following three are involved in their personalized immunotherapy and cancer vaccine strategy

Caperna LLC

Caperna

Caperna LLC is the fourth Moderna venture company — formed, funded and wholly-owned by Moderna — and focused exclusively on the advancement of personalized cancer vaccines.

Caperna will apply Moderna’s mRNA vaccine technology to the field of cancer vaccines, building on advances in recent years in cancer immunotherapy. Utilizing Moderna’s demonstrated engineering and process capability to synthesize over 1,000 unique novel mRNA’s per month in Moderna’s, automated, in-house productions systems. This provides the basis for a vision of rapid turnaround times that will allow Caperna’s personalized cancer vaccine, customized after tumor biopsy and sequencing to code for specific neoantigens in patients’ tumors, to be used to treat patients with aggressive tumors and high unmet need (rather than those with less aggressive tumors which can’t wait for prolonged turnaround times). Caperna will develop its personalized cancer vaccines in combination with checkpoint inhibitors that unleash the immune system and other cancer immunotherapies.

Corporate Facts

  • President: Tal Zaks, M.D., Ph.D.
  • Head of Research: Nicholas Valiante, Ph.D.
  • Head of Operations: Ted Ashburn, M.D., Ph.D.
  • Headquarters: 500 Technology Square, Cambridge, Mass.
  • Phone: 617-714-6500
  • Website: Caperna.com

 

Onkaido

Onkaido

Onkaido Therapeutics is the first Moderna venture company – formed, funded and wholly-owned by Moderna. Onkaido is focused exclusively on developing mRNA-based oncology treatments for currently undruggable targets or as a superior alternative to existing drug modalities. Onkaido is leveraging all of the tools and modalities developed at Moderna, with plans to rapidly turn mRNA science into truly novel cancer therapies that can make a real difference for patients.

Onkaido is currently focused on three therapeutic areas of oncology drug discovery and development: immuno-oncology, hepatocellular carcinoma (liver cancer) and myeloid malignancies – with programs investigating multiple targets and therapies simultaneously. Onkaido scientists are also exploring the power of mRNA technology in precision cancer pharmacology – researching areas such as tumor biology, targeting and gene silencing, driving the science toward the delivery of truly personalized cancer treatment.

Corporate Facts

  • President: Stephen Kelsey, M.D.
  • Headquarters: 500 Technology Square, Cambridge, Mass.
  • Phone: 617-714-6500
  • Website: Onkaido.com

 

Valera

Valera

Valera LLC is the second Moderna venture company — formed, funded and wholly-owned by Moderna — and focused exclusively on the advancement of vaccines and therapeutics for the prevention and treatment of viral, bacterial and parasitic infectious diseases.

The vaccines work of Valera builds on a body of preclinical research at Moderna showing the ability of modified mRNA to express viral antigens in vivo and to induce robust immune responses. Valera’s therapeutic passive immunity programs will expand on Moderna’s research using mRNA to express antibodies that bind to viral and other targets. The robust data from these programs across a range of preclinical infectious disease models, together with the inherent, rapid turn-around time in creating novel mRNA constructs, provide Valera with a potentially powerful and versatile new platform for the creation of a broad array of vaccines and passive immunity therapies.

Corporate Facts

  • President: Michael Watson, MB ChB, MRCP, AFPM
  • Chief Scientific OfficerGiuseppe Ciaramella, Ph.D.
  • Interim Chief Medical OfficerTal Zaks, M.D., Ph.D.
  • Headquarters: 500 Technology Square, Cambridge, Mass.
  • Phone: 617-714-6500
  • Website: Valeratx.com

And from http://endpts.com/neoantigens-beckon-merck-into-a-200m-cancer-collaboration-with-moderna/

Neoantigens beckon Merck into a $200M cancer collaboration with Moderna


Now that Galena has added fresh evidence that first-gen cancer vaccines make for a poor R&D program, Merck is betting $200 million upfront that the next-gen neoantigen approach to personalized cancer vaccines can succeed where all else has failed.

Merck is tying up with the mRNA specialists at Cambridge, MA-based Moderna, which has inked a long lineup of marquee partnerships. The big idea here is that each person’s cancer cells present unique “neoantigens” that can be used to tailor a cancer vaccine for each patient.

That’s a radical idea that has gained considerable steam in recent months, with Gritstone and Neon Therapeutics — paired now with Bristol-Myers on Opdivo — rounding up significant venture cash. Biotech billionaire Patrick Soon-Shiong has also jumped into the game, including it in its growing slate of cancer R&D work in a group of startups.

Moderna says it has already set up a manufacturing system that can be used to create these personalized vaccines in a matter of weeks. And Merck will use the partnership to advance new combination therapies that include its checkpoint inhibitor Keytruda.

The way the deal works, Moderna notes in its statement, is that Merck can step up after it sees some evidence in humans that the tech is working as planned. After human proof-of-concept, if Merck wants to opt in they can pay a significant milestone and then both companies can share the cost on Phase III and commercializations, profiting equally.Moderna CEO Stéphane Bancel says they can jump into the clinic next year.

The deal marks another rare pact by Merck R&D chief Roger Perlmutter, who’s been carefully focused on making Keytruda a foundation franchise that can sustain the company for years to come. While Merck has been a couple of steps behind Bristol-Myers in gaining market share, Perlmutter’s not settling for a second place finish.

“Combining immunotherapy with vaccine technology may be a new path toward improving outcomes for patients,” said Perlmutter, president, Merck Research Laboratories. “While the area of personalized cancer vaccine research has faced challenges in the past, there have been many recent advances, and we believe that working with Moderna to combine an immuno-oncology approach, using KEYTRUDA, with mRNA-based personalized cancer vaccines may have the potential to transform the treatment of cancer.”

From FierceBiotech on failure of Galena’s breast cancer vaccine trial

Galena plummets into microcap territory on Phase III breast cancer vaccine trial halt

Immunother Cancer. 2015; 3: 26.
Published online 2015 Jun 16. doi:  10.1186/s40425-015-0068-y
PMCID: PMC4468959

Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study

Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
CureVac GmbH, Paul-Ehrlich-Str. 15, Tuebingen, 72076 Germany
Charité University Hospital Berlin, Berlin, Germany
University Hospital Freiburg, Freiburg, Germany
Universitäty Hospital Essen, Essen, Germany
San Raffaele Scientific Institute, Milan, Italy
University Hospital of the Johannes-Gutenberg-University Mainz, Mainz, Germany
Ortenau Klinikum Offenburg-Gengenbach, Offenburg, Germany
University Hospital Göttingen, Göttingen/University Hospital Mannheim, Mannheim, Germany
University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany
Rippin-Consulting, Solingen, Germany
University Hospital Tuebingen, Tuebingen, Germany
Hubert Kübler, ed.nehcneum-ut.zrl@relbeuK.H.
corresponding authorCorresponding author.
#Contributed equally.

Abstract

Background

CV9103 is a prostate-cancer vaccine containing self-adjuvanted mRNA (RNActive®) encoding the antigens PSA, PSCA, PSMA, and STEAP1. This phase I/IIa study evaluated safety and immunogenicity of CV9103 in patients with advanced castration-resistant prostate-cancer.

Methods

44 Patients received up to 5 intra-dermal vaccinations. Three dose levels of total mRNA were tested in Phase I in cohorts of 3–6 patients to determine a recommended dose. In phase II, 32 additional patients were treated at the recommended dose. The primary endpoint was safety and tolerability, the secondary endpoint was induction of antigen specific immune responses monitored at baseline and at weeks 5, 9 and 17.

Results

The most frequent adverse events were grade 1/2 injection site erythema, injection site reactions, fatigue, pyrexia, chills and influenza-like illness. Possibly treatment related urinary retention occurred in 3 patients. The recommended dose was 1280 μg. A total of 26/33 evaluable patients treated at 1280 μg developed an immune response, directed against multiple antigens in 15 out of 33 patients. One patient showed a confirmed PSA response. In the subgroup of 36 metastatic patients, the Kaplan-Meier estimate of median overall survival was 31.4 months [95 % CI: 21.2; n.a].

Conclusions

The self-adjuvanted RNActive® vaccine CV9103 was well tolerated and immunogenic.

The technology is a versatile, fast and cost-effective platform allowing for creation of vaccines. The follow-up vaccine CV9104 including the additional antigens prostatic acid phosphatase (PAP) and Muc1 is currently being tested in a randomized phase IIb trial to assess the clinical benefit induced by this new vaccination approach.

SOURCE

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4468959/

Other articles in the Open Access Journal on Cancer Vaccines Include:

Cancer Vaccines: Targeting Cancer Genes for Immunotherapy – A Conference by Keystone Symposia on Molecular and Cellular Biology

AACR2016 – Cancer immunotherapy

Aduro Biotech Phase II Pancreatic Cancer Trial CRS-207 plus cancer vaccine GVAX Fails

Cases in Biotech Entrepreneurship: Selective Start Ups in 2016

at #JPM16 – Moderna Therapeutics turns away an extra $200 million: with AstraZeneca (collaboration) & with Merck ($100 million investment)

 

Read Full Post »

Personalized Immunotherapy: The Immuno-Oncology Summit August 30-31 2016 Boston MA

Reporter: Stephen J Williams, PhD

 

ANNOUNCEMENT

 

Leaders in Pharmaceutical Business intelligence (LPBI) Group will cover in Real Time using Social Media

The CHI’S 4TH ANNUAL IMMUNO-ONCOLOGY SUMMIT – Personalized Immunotherapy

Personalized Oncology in the Genomic Era: Expanding the Druggable Space

Aviva Lev-Ari, PhD, RN

will be streaming LIVE from the Marriott Long Wharf Hotel in Boston, MA

REGISTRATION

https://chidb.com/reg/imx/reg.asp

PROGRAM

http://www.immuno-oncologysummit.com/uploadedFiles/Immuno_Oncology_Summit/Agenda/16/2016-The-Immuno-Oncology-Summit-Brochure.pdf

 

 

Plenary Keynotes

TUESDAY | AUGUST 30

Matthew Goldstein

4:00 A New Era of Personalized Therapy: Using Tumor Neoantigens to Unlock the Immune System

Matthew J. Goldstein, M.D., Ph.D., Director, Translational Medicine, Neon Therapeutics, Inc.

Neon Therapeutics, Inc. launched in 2015 to focus on advancing neoantigen biology to improve cancer patient care. A neoantigen-based product engine will allow Neon to develop further treatment modalities including next-generation vaccines and T cell therapies targeting both personalized as well as shared neoantigens. The company’s first trial will launch later this year investigating the combination of a personalized, vaccine with nivolumab in advanced Melanoma, NSCLC, and Bladder Cancer.

Michael Rosenzweig

4:30 Emerging Innate Immune Targets for Enhancing Adaptive Anti-Tumor Responses

Michael Rosenzweig, Ph.D., Executive Director, Biology-Discovery, IMR Early Discovery, Merck Research Laboratories

Novel cancer immunotherapies targeting T cell checkpoint proteins have emerged as powerful tools to induce profound, durable regression and remission of many types of cancer. Despite these advances, multiple studies have demonstrated that not all patients respond to these therapies, and the ability to predict which patients may respond is limited. Harnessing the innate immune system to augment the adaptive anti-tumor response represents an attractive target for therapy, which has the potential to enhance both the percentage and rate of response to checkpoint blockade.

 

Morganna Freeman

5:00 Reading Tea Leaves:
The Dilemma of Prediction and Prognosis in Immunotherapy

Morganna Freeman, D.O., Associate Director, Melanoma & Cutaneous Oncology Program, The Angeles Clinic and Research Institute

With the rapid expansion of immunotherapeutics in oncology, scientifically significant advances have been made with both the depth and duration of antitumor responses. However, not all patients benefit, or quickly relapse, thus much scientific inquiry has been devoted to appropriate patient selection and how such obstacles might be overcome. While more is known about potential biomarkers, accurate prognostication persists as a knowledge gap, and efforts to bridge it will be discussed here.

Personalized Immunotherapy | The Immuno-Oncology Summit
August 30-31, 2016 | Marriott Long Wharf Hotel – Boston, MA

Personalized Immunotherapy
Personalized Oncology in the Genomic Era: Expanding the Druggable Space
August 30-31, 2016 | Learn More | Sponsorship & Exhibit Opportunities | Register by July 29 & SAVE up to $200!

Fueled with advances in genomic technologies, personalized oncology promises to innovate cancer therapy and target the previously undruggable space. Developments in immune checkpoint inhibitors, cancer vaccines, and adoptive T-cell therapies, as well as biomarker-driven immuno-oncology clinical trials, are enabling the next generation of cancer therapy. Cambridge Healthtech Institute’s Inaugural Personalized Immunotherapy meeting brings together clinical immuno-oncologists and thought leaders from pharmaceutical and biotech companies, and leading academic teams to share research and case studies in implementing patient-centric approaches to using the immune system to beat cancer.

TUMOR NEOANTIGENS FOR PERSONALIZED IMMUNOTHERAPY

Basics of Personalized Immunotherapy: What Is a Good Antigen?
Pramod K. Srivastava, M.D., Ph.D., Professor, Immunology and Medicine, Director, Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine

Novel Antibodies against Immunogenic Neoantigens
Philip M. Arlen, M.D., President & CEO, Precision Biologics, Inc.

PD-1 Blockade in Tumors with Mismatch-Repair Deficiency
Luis Alberto Diaz, M.D., Associate Professor, Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center

PERSONALIZED IMMUNOTHERAPY WITH CANCER VACCINES

Cancer Vaccines in the Era of Checkpoint Inhibitors
Keith L. Knutson, Ph.D., Professor, Immunology, Mayo Clinic

Developing Therapeutic Cancer Vaccine Strategies for Prostate Cancer
Ravi Madan, M.D., Clinical Director, Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health

Getting Very Personal: Fully Individualized Tumor Neoantigen-Based Vaccine Approaches to Cancer Therapy
Karin Jooss, Ph.D., CSO, Gritstone Oncology

Approaches to Assess Tumor Mutation Load for Selecting Patients for Cancer Immunotherapy
John Simmons, Ph.D., Manager, Research Services, Personal Genome Diagnostics

In situ Vaccination for Lymphoma
Joshua Brody, M.D., Director, Lymphoma Immunotherapy Program, Icahn School of Medicine at Mount Sinai

Immunotherapy Using Ad5 [E1-, E2b-] Vector Vaccines in the Cancer MoonShot 2020 Program
Frank R. Jones, Ph.D., Chairman & CEO, Etubics Corporation

PERSONALIZED CELL THERAPY

Integration of Natural Killer-Based Therapy into the Treatment of Lymphoma
Andrew M. Evens, D.O., Professor and Chief, Hematology/Oncology, Tufts University School of Medicine; Director, Tufts Cancer Center

Dendritic Cells: Personalized Cancer Vaccines and Inducers of Multi-Epitope-Specific T Cells for Adoptive Cell Therapy
Pawel Kalinski, M.D., Ph.D., Professor, Surgery, Immunology, and Bioengineering, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute

Mesothelin-Targeted CAR T-Cell Therapy for Solid Tumors
Prasad S. Adusumilli, M.D., FACS, Deputy Chief of Translational & Clinical Research, Thoracic Surgery, Memorial Sloan-Kettering Cancer Center

Synthetic Regulation of T Cell Therapies Adds Safety and Enhanced Efficacy to Previously Unpredicted Therapies
David M. Spencer, Ph.D., CSO, Bellicum Pharmaceuticals

Long-Term Relapse-Free Survival of Patients with Acute Myeloid Leukemia (AML) Receiving a Telomerase- Engineered Dendritic Cell Immunotherapy
Jane Lebkowski, Ph.D., President & CSO, Research and Development, Asterias Biotherapeutics

Activated and Exhausted Tumor Infiltrating B Cells in Non-Small Cell Lung Cancer Patients Present Antigen and Influence the Phenotype of CD4 Tumor Infiltrating T Cells
Tullia Bruno, Ph.D., Research Assistant Professor, Immunology, University of Pittsburgh

About the Immuno-Oncology Summit

CHI’s 4th Annual Immuno-Oncology Summit has been designed to support a coordinated effort by industry players to bring commercial immunotherapies and immunotherapy combinations through clinical development and into the market. This weeklong, nine-meeting set will include topics ranging from early discovery through clinical development as well as emerging areas such as oncolytic virotherapy. Overall, this event will provide a focused look at how researchers are applying new science and technology in the development of the next generation of effective and safe immunotherapies.

Monday, August 29 –
Tuesday, August 30
Tuesday, August 30 –
Wednesday, August 31
Thursday, September 1 –
Friday, September 2
Immunomodulatory Antibodies Combination Immunotherapy Adoptive T Cell Therapy
Oncolytic Virotherapy Personalized Immunotherapy Biomarkers for Immuno-Oncology
Training Seminar: Immunology for Drug Discovery Scientists Preclinical & Translational Immuno-Oncology Clinical Trials for Cancer Immunotherapy

For more info about sponsorship opportunities, including podium presentations and 1-2-1 meetings, please contact:

Companies A-K
Ilana Quigley
Sr Business Development Manager
781-972-5457
iquigley@healthtech.com
Companies L-Z:
Joe Vacca
Associate Director, Business Development
781-972-5431
jvacca@healthtech.com

For conference updates please visit
Immuno-OncologySummit.com/Personalized-Immunotherapy

Cambridge Healthtech Institute, 250 First Avenue, Suite 300, Needham, MA 02494 healthtech.com
Tel: 781-972-5400 | Fax: 781-972-5425

This email is being sent to sjwilliamspa@comcast.net. This email communication is for marketing purposes. If it is not of interest to you, please disregard and we apologize for any inconvenience this may have caused.
Visit www.chicorporate.com/corporate_unsubscribe.aspx to update usage.

Read Full Post »

Aduro Biotech Phase II Pancreatic Cancer Trial CRS-207 plus cancer vaccine GVAX Fails

Reporter: Stephen J. Williams, Ph.D

From Biospace News

May 16, 2016
By Alex Keown, BioSpace.com Breaking News Staff

BERKELEY, Calif. – Shares of Aduro Biotech (ADRO) have fallen more than 25 percent this morning following news that the company’s Phase II trial for its combination pancreatic cancer drug, CRS-207 did not meet its primary endpoint of survivability.

Aduro said its Eclipse trial of CRS-207 failed to show an improvement in overall survival for patients with pancreatic cancer who had failed at least two prior therapies in the metastatic setting. Median overall survival was 3.8 months for patients treated with the immunotherapy regimen of CRS-207 and the cancer vaccine GVAX pancreas, 5.4 months for patients treated with CRS-207 alone and 4.6 months for patients administered chemotherapy. Aduro said there were no reported safety concerns during the trial and full study findings will be presented at a later date.

Stephen T. Isaacs, chairman, president and chief executive officer of Aduro, called the findings a disappointing and “unexpected outcome.’

“While we are well aware of the very difficult-to-treat nature of late-stage metastatic pancreatic cancer, we are surprised by the divergence of these data from the results of our Phase IIa study. At the same time, we continue to look forward to the interim results later this year from our ongoing Stellar trial, which is evaluating CRS-207 and GVAX Pancreas with and without the anti-PD1 checkpoint inhibitor nivolumab as a second-line therapy for patients with metastatic pancreatic cancer,” Isaacs said in a statement.

For full story please see http://www.biospace.com/News/aduro-biotechs-stock-craters-after-pancreatic/419628/source=TopBreaking

Also from FierceBiotech

UPDATED: Aduro combo fails in a key pancreatic cancer study

Read Full Post »

Issues Need to be Resolved With Immuno-Modulatory Therapies: NK cells, mAbs, and adoptive T cells

Curator: Stephen J. Williams, PhD

nihms-618191-f0001NKvaciines

 

 

 

 

 

 

 

 

 

 

 

Immunotherapy. 2014;6(3):309-20. doi: 10.2217/imt.13.175.

Optimizing NKT cell ligands as vaccine adjuvants.

Carreño LJ1Kharkwal SSPorcelli SA.

Author information

Abstract

NKT cells are a subpopulation of T lymphocytes with phenotypic properties of both T and NK cells and a wide range of immune effector properties. In particular, one subset of these cells, known as invariant NKT cells (iNKT cells), has attracted substantial attention because of their ability to be specifically activated by glycolipid antigens presented by a cell surface protein called CD1d. The development of synthetic α-galactosylceramides as a family of powerful glycolipid agonists for iNKT cells has led to approaches for augmenting a wide variety of immune responses, including those involved in vaccination against infections and cancers. Here, we review basic, preclinical and clinical observations supporting approaches to improving immune responses through the use of iNKT cell-activating glycolipids. Results from preclinical animal studies and preliminary clinical studies in humans identify many promising applications for this approach in the development of vaccines and novel immunotherapies.

 

 

Cancer Res. 2013 Jul 1;73(13):3842-51. doi: 10.1158/0008-5472.CAN-12-1974. Epub 2013 May 23.

Avirulent Toxoplasma gondii generates therapeutic antitumor immunity by reversing immunosuppression in the ovarian cancer microenvironment.

Baird JR1Fox BASanders KLLizotte PHCubillos-Ruiz JRScarlett UKRutkowski MRConejo-Garcia JRFiering SBzik DJ.

Author information

Abstract

Reversing tumor-associated immunosuppression seems necessary to stimulate effective therapeutic immunity against lethal epithelial tumors. Here, we show this goal can be addressed using cps, an avirulent, nonreplicating uracil auxotroph strain of the parasite Toxoplasma gondii (T. gondii), which preferentially invades immunosuppressive CD11c(+) antigen-presenting cells in the ovarian carcinoma microenvironment. Tumor-associated CD11c(+) cells invaded by cps were converted to immunostimulatory phenotypes, which expressed increased levels of the T-cell receptor costimulatory molecules CD80 and CD86. In response to cps treatment of the immunosuppressive ovarian tumor environment, CD11c(+) cellsregained the ability to efficiently cross-present antigen and prime CD8(+) T-cell responses. Correspondingly, cps treatment markedly increased tumor antigen-specific responses by CD8(+) T cells. Adoptive transfer experiments showed that these antitumor T-cell responses were effective in suppressing solid tumor development. Indeed, intraperitoneal cps treatment triggered rejection of established ID8-VegfA tumors, an aggressive xenograft model of ovarian carcinoma, also conferring a survival benefit in a related aggressive model (ID8-Defb29/Vegf-A). The therapeutic benefit of cps treatment relied on expression of IL-12, but it was unexpectedly independent of MyD88 signaling as well as immune experience with T. gondii. Taken together, our results establish that cps preferentially invades tumor-associated antigen-presenting cells and restores their ability to trigger potent antitumor CD8(+) T-cell responses. Immunochemotherapeutic applications of cps might be broadly useful to reawaken natural immunity in the highly immunosuppressive microenvironment of most solid tumors.

 

Oncoimmunology. 2013 Jun 1;2(6):e24677. Epub 2013 Apr 29.

TLR3 agonists improve the immunostimulatory potential of cetuximab against EGFR+ head and neck cancer cells.

Ming Lim C1Stephenson RSalazar AMFerris RL.

Author information

Abstract

Toll-like receptor 3 (TLR3) agonists have been extensively used as adjuvants for anticancer vaccines. However, their immunostimulatory effects and precise mechanisms of action in the presence of antineoplastic monoclonal antibodies (mAbs) have not yet been evaluated. We investigated the effect of TLR3 agonists on cetuximab-mediated antibody-dependent cellular cytotoxicity (ADCC) against head and neck cancer (HNC) cells, as well as on dendritic cell (DC) maturation and cross-priming of epidermal growth factor receptor (EGFR)-specific CD8+ T cells. The cytotoxic activity of peripheral blood mononuclear cells (PBMCs) or isolated natural killer (NK) cells expressing polymorphic variants (at codon 158) of the Fcγ receptor IIIa (FcγIIIa) was determined in 51Cr release assays upon incubation with the TLR3 agonist poly-ICLC. NK cell stimulation was measured based on activation and degranulation markers, while DC maturation in the presence of poly-ICLC was assessed using flow cytometry. The DC-mediated cross priming of EGFR-specific CD8+ T cells was monitored upon in vitro stimulation with tetramer-based flow cytometry. TLR3-stimulated, unfractionated PBMCs from HNC patients mediated robust cetuximab-dependent ADCC, which was abrogated by NK-cell depletion. The cytolytic activity of TLR3-stimulated NK cells differed among cells expressing different polymorphic variants of FcγRIIIa, and NK cells exposed to both poly-ICLC and cetuximab expressed higher levels of CD107a and granzyme B than their counterparts exposed to either stimulus alone. Poly-ICLC plus cetuximab also induced a robust upregulation of CD80, CD83 and CD86 on the surface of DCs, a process that was partially NK-cell dependent. Furthermore, DCs matured in these conditions exhibited improved cross-priming abilities, resulting in higher numbers of EGFR-specific CD8+ T cells. These findings suggest that TLR3 agonists may provide a convenient means to improve the efficacy of mAb-based anticancer regimens.

Ann Oncol. 2012 Sep; 23(Suppl 8): viii6–viii9.

doi:  10.1093/annonc/mds256

PMCID: PMC4085883

Immuno-oncology: understanding the function and dysfunction of the immune system in cancer

  1. J. Finn*

Interactions between the Immune System and Cancer

Evidence has been accumulating since the middle of the last century, first from animal models and later from studies in cancer patients, that the immune system can recognise and reject tumours. The goal of tumour immunology has been to understand the components of the immune system that are important for tumour immunosurveillance and tumour rejection to understand how, when, and why they fail in cases of clinical disease. Immunotherapy, which involves strengthening the cancer patient’s immune system by improving its ability to recognise the tumour or providing a missing immune effector function, is one treatment approach that holds promise of a life-long cure [4].

Studies of cancer–immune system interactions have revealed that every known innate and adaptive immune effector mechanism participates in tumour recognition and control [5]. The first few transformed cells are detected by NK cells through their encounter with specific ligands on tumour cells. This leads to the destruction of some transformed cells and the uptake and processing of their fragments by macrophages and dendritic cells. In turn, these macrophages and dendritic cells are activated to secrete many inflammatory cytokines and present tumour cell-derived molecules to T- and B cells. Activation of T- and B cells leads to the production of additional cytokines that further promote activation of innate immunity and support the expansion and production of tumour-specific T cells and antibodies, respectively. The full power of the adaptive immune system leads to the elimination of remaining tumour cells and, importantly, to the generation of immune memory to specific tumour components that will serve to prevent tumour recurrence.

Effectors of adaptive immunity, such as CD4+ helper T cells, CD8+ cytotoxic T cells, and antibodies, specifically target tumour antigens; i.e. molecules expressed in tumour cells, but not in normal cells. Tumour antigens are normal cellular proteins that are abnormally expressed as a result of genetic mutations, quantitative differences in expression, or differences in posttranslational modifications [5]. In tumour types that have a well-documented viral origin, such as cervical cancer, caused by the human papillomavirus [5], or hepatocellular carcinoma caused by the hepatitis B virus [6], viral proteins can also serve as tumour antigens and targets for antitumour immune response [7].

The first indication that tumours carried molecules distinct from those on the normal cell of origin was derived from immunising mice with human tumours and selecting antibodies that recognised human tumour cells but not their normal counterparts. The major question was whether some, or all, of these molecules would also be recognised by the human immune system. 2011 was an important anniversary for human tumour immunology, marking 20 years since the publication by van der Bruggen et al. [8] that described the cloning of MAGE-1, a gene that encodes a human melanoma antigen recognised by patient’s antitumour T cells. This was not a mutant protein; its recognition by the immune system was due to the fact that it was only expressed by transformed, malignant cells and, with the exception of testicular germ cells, was not expressed in normal adult tissue. Many similar discoveries followed, with each new molecule providing a better understanding of what might be good targets for different forms of cancer immunotherapy. Tumour antigens have been tested as vaccines, as targets for monoclonal antibodies, and as targets for adoptively transferred cytotoxic T cells. There is a wealth of publications from preclinical studies targeting these antigens and results from phase I/II clinical trials. Recently, these studies were critically reviewed and a list of tumour antigens with the largest body of available data compiled [9]. The goal was to encourage faster progress in the design, testing, and approval of immunotherapeutic reagents that incorporate or target the most promising antigens.

 

As highlighted in the article two scenarios which present problems emerged:

  1. In the past, immunotherapy was referred to as ‘passive’ (e.g. the infusion of preformed immune effectors, such as antibodies, cytokines, or activated T cells, NK cells, or lymphokine-activated killer cells), presumably acting directly on the tumour and independent of the immune system or ‘active’ (e.g. vaccines), designed to activate and therefore be dependent on the patient’s immune system. it has since become clear that both passive and active immunotherapies depend on the patient’s immune system for long-term tumour control or complete tumour elimination. anticancer monoclonal antibodies are a well-established class of immunotherapeutic agent. HOWEVER, The potential of these antibodies is drastically undermined by their administration relatively late in the disease course, when the patient’s immune system is largely compromised. Under more optimal conditions, antibody treatment might result not only in the direct cytostatic or cytotoxic effect on the tumour cell, but also in the loading of antibody-bound tumour antigens onto antigen presenting cells (APC) in the tumour microenvironment. The resultant cross-presentation to antitumour T- and B cells could result in additional antibodies to these antigens being produced, and propagation of the immune response at the tumour site would maintain tumour elimination long after the infused monoclonal antibody is gone.
  2. The same scenario could be predicted for adoptively transferred T cells. Unlike antibodies, transferred T cells persist longer and may provide a memory response [14]; however, as long as the memory response is restricted to one clone, or a limited number of clones, then antigen-negative tumours will be able to escape. In addition, cancer vaccines encounter large numbers of immunosuppressive Tregand MDSC in circulation, as well as immunosuppressive cell-derived soluble products that flood the lymph nodes, preventing maturation of APCs and activation of T cells. Even when vaccines are delivered in the context of ex vivo matured and activated dendritic cells, their ability to activate T cells is compromised by the high-level expression of various molecules on T cells that block this process.

The scenarios proposed above present a rather bleak picture of the potential of immunotherapy to achieve the cure for cancer that has eluded standard therapy [15]. Interestingly, failures of some standard therapies are beginning to be ascribed to their inability to activate the patient’s immune system [16]. However, rather than seeing the picture as a deterrent, it should be considered as a road map, providing at least two major directions for new developments in immunotherapy.

The first direction is to continue using the old classes of immunotherapy that target the cancer directly, but to use them in combination with therapies that target the immune system in the tumour microenvironment, such as cytokines, suppressors of Treg or MDSC activity, or antibodies that modulate T-cell activity. The recently approved antibody, ipilimumab, which acts to sustain cytotoxic T-cell activity by augmenting T-cell activation and proliferation, is one example of such an immunomodulatory agent [17].

The other direction is to use immunotherapies, both old and new, for preventing cancer in individuals at high risk [18]. Studies of the tumour microenvironment are providing information about immunosurveillance of tumours from early premalignant lesions to more advanced dysplastic lesions to cancer. At each step, tumour-derived and immune system-derived components have a unique composition that will have distinct effects on immunotherapy. Because these premalignant microenvironments are less developed and immunosuppression is less entrenched, it should be easier to modulate towards the elimination of abnormal cells.

 

Cancer Immunol Immunother. 2011 Sep;60(9):1309-17. doi: 10.1007/s00262-011-1038-y. Epub 2011 May 28.

Tumor immunotherapy using adenovirus vaccines in combination with intratumoral doses of CpG ODN.

Geary SM1Lemke CDLubaroff DMSalem AK.

Author information

Abstract

The combination of viral vaccination with intratumoral (IT) administration of CpG ODNs is yet to be investigated as an immunotherapeutic treatment for solid tumors. Here, we show that such a treatment regime can benefit survival of tumor-challenged mice. C57BL/6 mice bearing ovalbumin (OVA)-expressing EG.7 thymoma tumors were therapeutically vaccinated with adenovirus type 5 encoding OVA (Ad5-OVA), and the tumors subsequently injected with the immunostimulatory TLR9 agonist, CpG-B ODN 1826 (CpG), 4, 7, 10, and 13 days later. This therapeutic combination resulted in enhanced mean survival times that were more than 3.5× longer than naïve mice, and greater than 40% of mice were cured and capable of resisting subsequent tumor challenge. This suggests that an adaptive immune response was generated. Both Ad5-OVA and Ad5-OVA + CpG IT treatments led to significantly increased levels of H-2 K(b)-OVA-specific CD8+ lymphocytes in the peripheral blood and intratumorally. Lymphocyte depletion studies performed in vivo implicated both NK cells and CD8+ lymphocytes as co-contributors to the therapeutic effect. Analysis of tumor infiltrating lymphocytes (TILs) on day 12 post-tumor challenge revealed that mice treated with Ad5-OVA + CpG IT possessed a significantly reduced percentage of regulatory T lymphocytes (Tregs) within the CD4+ lymphocyte population, compared with TILs isolated from mice treated with Ad5-OVA only. In addition, the proportion of CD8+ TILs that were OVA-specific was reproducibly higher in the mice treated with Ad5-OVA + CpG IT compared with other treatment groups. These findings highlight the therapeutic potential of combining intratumoral CpG and vaccination with virus encoding tumor antigen.

 

Adv Drug Deliv Rev. 2009 Mar 28;61(3):268-74. doi: 10.1016/j.addr.2008.12.005. Epub 2009 Jan 7.

CpG oligonucleotide as an adjuvant for the treatment of prostate cancer.

Lubaroff DM1Karan D.

Author information

Abstract

The use of an adenovirus transduced to express a prostate cancer antigen (PSA) as a vaccine for the treatment of prostate cancer has been shown to be active in the destruction of antigen-expressing prostate tumor cells in a pre-clinical model, using Balb/C or PSA transgenic mice. The destruction of PSA-secreting mouse prostate tumors was observed in Ad/PSA immunized mice in a prophylaxis study with 70% of the mice surviving long term tumor free. This successful immunotherapy was not observed in therapeutic studies in which tumors were established before vaccination and the development of anti-PSA immune response was not as easily generated in PSA transgenic mice. Immunization of conventional and transgenic animals was enhanced by incorporating a collagen matrix into the immunizing injection. Therefore the need to strengthen anti-PSA and anti-prostate cancer immunity was an obvious next step in developing a successful prostate cancer immunotherapy. Because the use ofimmunostimulatory CpG motifs was shown to enhance immune responses to a wide variety of antigens, our studies incorporated CpG into the Ad/PSA vaccine experimental plans. The results of the subsequent studies demonstrated a dichotomy where Ad/PSA plus CpG enhanced the in vivo destruction of PSA-secreting tumors and the survival of experimental animals, but revealed that the number and in vitro activities of antigen specific CD8+ T cells was decreased as compared to the values observed when the vaccine alone was used for immunization. The dichotomous observations were confirmed using another antigen system, OVA also incorporated into a replication defective adenovirus. Despite the reduction in antigen-specific CD8+ cells after vaccine plus CpG immunization the enhanced destruction of sc and systemic tumors was shown to be mediated entirely by CD8+ T cells. Finally, the reduction of the CD8+ T cells was the result of an observed decrease in the proliferation of the antigen specific cell population.

J Invest Dermatol. 2004 Aug;123(2):371-9.

 

CpG motifs are efficient adjuvants for DNA cancer vaccines.

Schneeberger A1Wagner CZemann ALührs PKutil RGoos MStingl GWagner SN.

Author information

Abstract

DNA vaccines can induce impressive specific cellular immune response (IR) when taking advantage of their recognition as pathogen-associated molecular patterns (PAMP) through Toll-like receptors (TLR) expressed on/in cells of the innate immune system. Among the many types of PAMP,immunostimulatory DNA, so-called CpG motifs, was shown to interact specifically with TLR9, which is expressed in plasmacytoid dendritic cells(pDC), a key regulatory cell for the activation of innate and adaptive IR. We now report that CpG motifs, when introduced into the backbone, are a useful adjuvant for plasmid-based DNA (pDNA) vaccines to induce melanoma antigen-specific protective T cell responses in the Cloudman M3/DBA/2 model. The CpG-enriched pDNA vaccine induced protection against subsequent challenge with melanoma cells at significantly higher levels than its parental unmodified vector. Preferential induction of an antigen-specific, protective T cell response could be demonstrated by (i) induction of antigen-dependent tumor cell protection, (ii) complete loss of protection by in vivo CD4+/CD8+T cell- but not NK cell-depletion, and (iii) the detection of antigen-specific T cell responses but not of relevant NK cell activity in vitro. These results demonstrate that employing PAMP in pDNA vaccines improves the induction of protective, antigen-specific, T cell-mediated IR.

 

J Biomed Sci. 2016 Jan 25;23(1):16. doi: 10.1186/s12929-016-0238-3.

Combination of the toll like receptor agonist and α-Galactosylceramide as an efficient adjuvant for cancer vaccine.

Gableh F1Saeidi M2Hemati S3Hamdi K4Soleimanjahi H5Gorji A6,7,8Ghaemi A9,10,11.

Author information

Abstract

BACKGROUND:

DNA vaccines have emerged as an attractive approach for the generation of cytotoxic T lymphocytes (CTL). In our previous study, we found That Toll like receptor (TLR) ligands are promising candidates for the development of novel adjuvants for DNA vaccine. To improve the efficacy of DNA vaccine directed against human papillomavirus (HPV) tumors, we evaluated whether co-administration of a TLR4 ligand, monophosphoryl lipid A (MPL), and Natural Killer T Cell Ligand α-Galactosylceramide(α-GalCer) adjuvants with DNA vaccine would influence the anti-tumor efficacy of DNA vaccinations.

METHODS:

We investigated the effectiveness of α-GalCer and MPL combination as an adjuvant with an HPV-16 E7 DNA vaccine to enhance antitumor immune responses.

RESULTS:

By using adjuvant combination for a DNA vaccine, we found that the levels of lymphocyte proliferation, CTL activity, IFN- γ, IL-4 and IL-12 responses, and tumor protection against TC-1 cells were significantly increased compared to the DNA vaccine with individual adjuvants. In addition, inhibition of IL-18 signaling during vaccination decreased IFN-γ responses and tumor protection, and that this inhibition suggested stimulatory role of IL-18 in adjuvant effects of α-GalCer and MPL combination.

CONCLUSION:

The strong adjuvanticity associated with α-GalCer/MPL combination may to be an important tool in the development of novel and strong cancer immunotherapy.

Cancer Sci. 2015 Dec;106(12):1659-68. doi: 10.1111/cas.12824. Epub 2015 Nov 18.

Adjuvant for vaccine immunotherapy of cancer – focusing on Toll-like receptor 2 and 3 agonists for safely enhancing antitumor immunity.

Seya T1Shime H1Takeda Y1Tatematsu M1Takashima K1Matsumoto M1.

Author information

Abstract

Immune-enhancing adjuvants usually targets antigen (Ag)-presenting cells to tune up cellular and humoral immunity. CD141(+) dendritic cells (DC) represent the professional Ag-presenting cells in humans. In response to microbial pattern molecules, these DCs upgrade the maturation stage sufficient to improve cross-presentation of exogenous Ag, and upregulation of MHC and costimulators, allowing CD4/CD8 T cells to proliferate and liberating cytokines/chemokines that support lymphocyte attraction and survival. These DCs also facilitate natural killer-mediated cell damage. Toll-like receptors (TLRs) and their signaling pathways in DCs play a pivotal role in DC maturation. Therefore, providing adjuvants in addition to Ag is indispensable for successful vaccine immunotherapy for cancer, which has been approved in comparison with antimicrobial vaccines. Mouse CD8α(+) DCs express TLR7 and TLR9 in addition to the TLR2 family (TLR1, 2, and 6) and TLR3, whereas human CD141(+) DCs exclusively express the TLR2 family and TLR3. Although human and mouse plasmacytoid DCs commonly express TLR7/9 to respond to their agonists, the results on mouse adjuvant studies using TLR7/9 agonists cannot be simply extrapolated to human adjuvant immunotherapy. In contrast, TLR2 and TLR3 are similarly expressed in both human and mouse Ag-presenting DCs. Bacillus Calmette-Guerin peptidoglycan and polyinosinic-polycytidylic acid are representative agonists for TLR2 and TLR3, respectively, although they additionally stimulate cytoplasmic sensors: their functional specificities may not be limited to the relevant TLRs. These adjuvants have been posted up to a certain achievement in immunotherapy in some cancers. We herein summarize the history and perspectives of TLR2 and TLR3 agonists in vaccine-adjuvant immunotherapy for cancer.

Adv Exp Med Biol. 2015;850:81-91. doi: 10.1007/978-3-319-15774-0_7.

Molecular Programming of Immunological Memory in Natural Killer Cells.

Beaulieu AM1Madera SSun JC.

Author information

Abstract

Immunological memory is a hallmark of the adaptive immune system. Although natural killer (NK) cells have traditionally been classified as a component of the innate immune system, they have recently been shown in mice and humans to exhibit certain features of immunological memory, including an ability to undergo a clonal-like expansion during virus infection, generate long-lived progeny (i.e. memory cells), and mediate recall responses against previously encountered pathogens–all characteristics previously ascribed only to adaptive immune responses by B and T cells in mammals. To date, the molecular events that govern the generation of NK cell memory are not completely understood. Using a mouse model of cytomegalovirus infection, we demonstrate that individual pro-inflammatory IL-12, IL-18, and type I-IFN signaling pathways are indispensible and play non-redundant roles in the generation of virus-specific NK cell memory. Furthermore, we discovered that antigen-specific proliferation and protection by NK cells is mediated by the transcription factor Zbtb32, which is induced by pro-inflammatory cytokines and promotes a cell cycle program in activated NK cells. A greater understanding of the molecular mechanisms controlling NK cell responses will provide novel strategies for tailoring vaccines to target infectious disease.

 

Read Full Post »

Meeting Announcement: Cancer Immunotherapy and Combinations June 15-16 2016

Reporter: Stephen J. Williams, PhD

 

Cancer Immunotherapy & Combinations – June 15-16, 2016 in Boston, MA

YouTubeLinkedInTwitter#CHIWPC16

Final Brochure PDF | Learn More | Sponsorship & Exhibit Details | Register by March 4 & SAVE up to $400!

Cambridge Healthtech Institute’s inaugural Cancer Immunotherapy and Combinations meeting will convene immuno-oncology researchers, cancer immunotherapy developers, and technology providers to discuss next-generation approaches and combinations, including small molecule development, to enhance the efficacy of checkpoint inhibitors.

BISPECIFIC ANTIBODIES – DUAL TARGETING

FEATURED PRESENTATION: ANTI-PD1 OR CD137 ENHANCES NK-CELL CYTOTOXICITY TOWARDS CD30+ HODGKIN LYMPHOMA INDUCED BY CD30/CD16A TANDAB AFM13
Martin Treder, Ph.D., CSO, R&D, Affimed

In vivo Efficacy of Bispecific Antibodies Targeting Two Immune-Modulatory Receptors
Jacqueline Doody, Ph.D., Vice President, Immunology, F-star Biotechnology, Ltd

Dual-Targeting Bispecific Antibodies for Selective Neutralization of CD47 on Cancer Cells
Krzysztof Masternak, Ph.D., Head, Biology, Therapeutic Antibody Discovery, Novimmune

Update on MCLA-134: A Biclonics® Binding Two Immunomodulatory Targets Expressed by T Cells
Mark Throsby, Ph.D., CSO, Merus

The ImmTAC Technology: A Cutting-Edge Immunotherapy for Cancer Treatment
Samir Hassan, Ph.D., Director, Translational Research & Development, Immunocore Ltd.

RADIOTHERAPY AND CHEMOTHERAPY – PD-1 COMBINATIONS

Rational Development of Combinations of Antiangiogenic Therapy with Immune Checkpoint Blockers Using Mouse Models of HCC and Cirrhosis
Dan Duda, D.M.D., Ph.D., Associate Professor, Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School

Harnessing the Immune Microenvironment of Gastrointestinal Cancers Using Combined Modalities
Osama Rahma, M.D., Assistant Professor, Internal Medicine/Oncology, University of Virginia

AGONIST – PD-1 AND CTLA-4 COMBINATIONS

The Role of the Target in the Disposition and Immunogenicity of an Anti-GITR Agonist Antibody
Enrique Escandón, Ph.D., Senior Principal Scientist, DMPK and Disposition, Merck

Combination of 4-1BB Agonist and PD-1 Antagonist Promotes Antitumor Effector/Memory CD8 T Cells
Changyu Wang, Ph.D., Director, Cancer Immunology, Pfizer

Combination Immunotherapy with Checkpoint Blockade, Agonist Anti-OX40 mAb, and Vaccination Rescues Anergic CD8 T Cells
William Redmond, Ph.D., Associate Member, Laboratory of Cancer Immunotherapy, Earle A. Chiles Research Institute, Providence Portland Medical Center

Interactive Breakout Discussion Groups with Continental Breakfast

This session features various discussion groups that are led by a moderator/s who ensures focused conversations around the key issues listed. Attendees choose to join a specific group and the small, informal setting facilitates sharing of ideas and active networking. Continental breakfast is available for all participants.

Topic: Small Molecule Targeting of IDO1 and TDO for Cancer Immunotherapy

Moderator: Rogier Buijsman, Ph.D., Head, Chemistry, Netherlands Translational Research Center B.V. (NTRC)

  • Overcoming challenges of current IDO1 inhibitors lacking selectivity over TDO and having suboptimal drug-like properties
  • Advances in IDO1 and TDO inhibitor screening
  • Is selective IDO1 or TDO inhibitors is required, or a dual IDO1/TDO inhibitor is preferred to obtain optimal efficacy and safety in the clinic?

Topic: Strategies for Developing Bispecific Antibodies for Cancer Immunotherapy

Moderator: Krzysztof Masternak, Ph.D., Head, Biology, Therapeutic Antibody Discovery, Novimmune

  • Considerations for efficacy in vitro and in vivo: selectivity for tumor cells, ADCP, ADCC, in vivo efficacy (xenograft models)
  • Insights into mechanisms of action
  • Safety considerations: binding selectivity, PK and tox studies

Topic: Combining Standard Antiangiogenic Therapy with Immune Checkpoint Inhibitors

Moderator: Dan Duda, D.M.D., Ph.D., Associate Professor, Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School

  • Will checkpoint combination with chemotherapy or other targeted agents prove to have too many toxicity issues?
  • How do we minimize overlapping toxic effects of radiation and immunotherapy?
  • How to optimize the sequencing of these two treatment modalities?

SMALL MOLECULE INHIBITORS AS SINGLE AND CHECKPOINT COMBINATION AGENTS

Selective Small Molecule Inhibitors of IDO1 and TDO for Cancer Immunotherapy
Rogier Buijsman, Ph.D., Head, Chemistry, Netherlands Translational Research Center B.V. (NTRC)

Potent and Selective Small Molecule USP7 Inhibitors for Cancer Immunotherapy
Suresh Kumar, Ph.D., Director, R&D, Progenra, Inc.

Epigenetic Agents for Combination with Cancer Immunotherapy
Svetlana Hamm, Ph.D., Head, Biology, Translational Pharmacology, 4SC Group

VACCINES AND CHECKPOINT BLOCKADE IMMUNOTHERAPY

Immunotherapy for Mesothelioma with an in vivo DC Vaccine and PD-1/PD-L1 Blockade
Huabiao Chen, M.D., Ph.D., Principal Investigator, Vaccine and Immunotherapy Center, Massachusetts General Hospital

Bringing Together Checkpoint Inhibition with Vaccines Using Customizing Capsids
Willie Quinn, Ph.D., President & CEO, Bullet Bio

Recommended All Access Package:

June 14 SC1: Immunosequencing: Generating a New Class of Cancer Immunotherapy Diagnostics*

June 14 SC5: Convergence of Immunotherapy and Epigenetics for Cancer Treatment*

June 14 SC8: Rational Design of Cancer Combination Therapies*

June 15-16: Cancer Immunotherapy and Combinations

June 16-17: Tumor Models for Cancer Immunotherapy

* Separate registration required.

Exhibit booth space has sold out! The few remaining spaces are being sold via sponsorship only. To customize yoursponsorship package, please contact:
Joseph Vacca, M.Sc., Associate Director, Business Development, 781-972-5431, jvacca@healthtech.com

For more information visit

WorldPreclinicalCongress.com/Cancer-Immunotherapy-Combinations

Cambridge Healthtech Institute, 250 First Avenue, Suite 300, Needham, MA 02494 healthtech.com
Tel: 781-972-5400 | Fax: 781-972-5425

This email is being sent to sjwilliamspa@comcast.net. This email communication is for marketing purposes. If it is not of interest to you, please disregard and we apologize for any inconvenience this may have caused.
Visit www.chicorporate.com/corporate_unsubscribe.aspx to update usage.

Read Full Post »

%d bloggers like this: