Immunology and Biotherapies
37.9K views | +0 today
Immunology and Biotherapies
Page Ressources et Actualités du DIU immunologie et biothérapies
Your new post is loading...
Your new post is loading...
Rescooped by Gilbert C FAURE from Cancer Vaccines Collection
Scoop.it!

The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma

Findings

The anti-PD-1 and anti-PD-L1 agents have been reported to have impressive antitumor effects in several malignancies, including melanoma. The greatest clinical activity in unselected patients has been seen in melanoma. Tumor expression of PD-L1 is a suggestive, but inadequate, biomarker predictive of response to immune-checkpoint blockade. However, tumors expressing little or no PD-L1 are less likely to respond to PD-1 pathway blockade. Combination checkpoint blockade with PD-1 plus cytotoxic T-lymphocyte antigen (CTLA)-4 blockade appears to improve response rates in patients who are less likely to respond to single-checkpoint blockade. Toxicity with PD-1 blocking agents is less than the toxicity with previous immunotherapies (eg, interleukin 2, CTLA-4 blockade). Certain adverse events can be severe and potentially life threatening, but most can be prevented or reversed with close monitoring and appropriate management.

Implications

This family of immune-checkpoint inhibitors benefits not only patients with metastatic melanoma but also those with historically less responsive tumor types. Although a subset of patients responds to single-agent blockade, the initial trial of checkpoint-inhibitor combinations has reported a potential to improve response rates. Combination therapies appear to be a means of increasing response rates, albeit with increased immune-related adverse events. As these treatments become available to patients, education regarding the recognition and management of immune-related effects of immune-checkpoint blockade will be essential for maximizing clinical benefit.


Via Krishan Maggon
Krishan Maggon 's curator insight, May 15, 2015 4:02 AM

doi:10.1016/j.clinthera.2015.02.018

 

Clinical Therapeutics

Volume 37, Issue 4, 1 April 2015, Pages 764–782

Review Article The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in MelanomaKathleen M. Mahoney, MD, PhD1, 2, , , Gordon J. Freeman, PhD2, David F. McDermott, MD1

Rescooped by Gilbert C FAURE from Cancer Vaccines Collection
Scoop.it!

A Novel Therapy for Melanoma Developed in Mice: Transformation of Melanoma into Dendritic Cells with Listeria monocytogenes

A Novel Therapy for Melanoma Developed in Mice: Transformation of Melanoma into Dendritic Cells with  Listeria monocytogenes | Immunology and Biotherapies | Scoop.it
Listeria monocytogenes is a gram-positive bacteria and human pathogen widely used in cancer immunotherapy because of its capacity to induce a specific cytotoxic T cell response in tumours.

Via Krishan Maggon
Krishan Maggon 's curator insight, March 12, 2015 3:53 AM

These results support the use of low doses of pathogenic Listeri as safe melanoma therapeutic vaccines that do not require antibiotics for bacterial removal. 


Citation: Bronchalo-Vicente L, Rodriguez-Del Rio E, Freire J, Calderon-Gonzalez R, Frande-Cabanes E, et al. (2015) A Novel Therapy for Melanoma Developed in Mice: Transformation of Melanoma into Dendritic Cells with Listeria monocytogenes. PLoS ONE 10(3): e0117923. doi:10.1371/journal.pone.0117923

Rescooped by Gilbert C FAURE from Cancer Vaccines Collection
Scoop.it!

Cancer Gene Therapy - Expansion of melanoma-specific lymphocytes in alternate gamma chain cytokines: gene expression variances between T cells and T-cell subsets exposed to IL-2 versus IL-7/15

Cancer Gene Therapy - Expansion of melanoma-specific lymphocytes in alternate gamma chain cytokines: gene expression variances between T cells and T-cell subsets exposed to IL-2 versus IL-7/15 | Immunology and Biotherapies | Scoop.it
Cancer Gene Therapy is the essential gene therapy resource for cancer researchers and clinicians, keeping readers up to date with the latest developments in gene therapy for cancer.

Via Krishan Maggon
Krishan Maggon 's curator insight, November 15, 2014 9:07 AM

OA


Cancer Gene Therapy (2014) 21, 441–447; doi:10.1038/cgt.2014.48; published online 19 September 2014

Expansion of melanoma-specific lymphocytes in alternate gamma chain cytokines: gene expression variances between T cells and T-cell subsets exposed to IL-2 versus IL-7/15

C K Zoon1, E Seitelman2, S Keller3, L Graham2, T L Blevins4, C I Dumur4 and H D Bear1,2

1Department of Surgery, Virginia Commonwealth University Health System, Richmond, VA, USA2Division of Surgical Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA3Department of Microbiology and Immunology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA4Department of Pathology, Virginia Commonwealth University Health System, Richmond, VA, USA

Correspondence: CK Zoon, Dr CK Zoon, Department of Surgery, Virginia Commonwealth University Health System, Medical College of Virginia Campus, PO Box 980135, Richmond, VA 23298, USA. E-mail:ckzoon@comcast.net

Received 16 June 2014; Revised 7 August 2014; Accepted 8 August 2014
Advance online publication 19 September 2014

Rescooped by Gilbert C FAURE from Cancer Vaccines Collection
Scoop.it!

Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients

Abstract

Enhancing immune responses with immune-modulatory monoclonal antibodies directed to inhibitory immune receptors is a promising modality in cancer therapy. Clinical efficacy has been demonstrated with antibodies blocking inhibitory immune checkpoints such as cytotoxic T lymphocyte–associated antigen 4 (CTLA-4) or PD-1/PD-L1. Treatment with ipilimumab, a fully human CTLA-4–specific mAb, showed durable clinical efficacy in metastatic melanoma; its mechanism of action is, however, only partially understood. This is a study of 29 patients with advanced cutaneous melanoma treated with ipilimumab. We analyzed peripheral blood mononuclear cells (PBMCs) and matched melanoma metastases from 15 patients responding and 14 not responding to ipilimumab by multicolor flow cytometry, antibody-dependent cell-mediated cytotoxicity (ADCC) assay, and immunohistochemistry. PBMCs and matched tumor biopsies were collected 24 h before (i.e., baseline) and up to 4 wk after ipilimumab. Our findings show, to our knowledge for the first time, that ipilimumab can engage ex vivo FcγRIIIA (CD16)-expressing, nonclassical monocytes resulting in ADCC-mediated lysis of regulatory T cells (Tregs). In contrast, classical CD14++CD16−monocytes are unable to do so. Moreover, we show that patients responding to ipilimumab display significantly higher baseline peripheral frequencies of nonclassical monocytes compared with nonresponder patients. In the tumor microenvironment, responders have higher CD68+/CD163+ macrophage ratios at baseline and show decreased Treg infiltration after treatment. Together, our results suggest that anti–CTLA-4 therapy may target Tregs in vivo. Larger translational studies are, however, warranted to substantiate this mechanism of action of ipilimumab in patients.


Via Krishan Maggon
Krishan Maggon 's curator insight, May 10, 2015 1:55 PM

PNAS

Emanuela Romano, doi: 10.1073/pnas.1417320112

 

Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patientsEmanuela Romanoa,b,c,1, Monika Kusio-Kobialkab, Periklis G. Foukasc,d, Petra Baumgaertnerc,Christiane Meyerc, Pierluigi Ballabenie, Olivier Michielina,c, Benjamin Weidef, Pedro Romeroc, andDaniel E. Speiserc

Author Affiliations

Edited by Ira Mellman, Genentech, Inc., South San Francisco, CA, and approved March 30, 2015 (received for review September 9, 2014)

Krishan Maggon 's curator insight, May 12, 2015 3:05 AM

PNAS

Emanuela Romano, doi: 10.1073/pnas.1417320112

 

Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patientsEmanuela Romanoa,b,c,1, Monika Kusio-Kobialkab, Periklis G. Foukasc,d, Petra Baumgaertnerc,Christiane Meyerc, Pierluigi Ballabenie, Olivier Michielina,c, Benjamin Weidef, Pedro Romeroc, andDaniel E. Speiserc

Author Affiliations

Edited by Ira Mellman, Genentech, Inc., South San Francisco, CA, and approved March 30, 2015 (received for review September 9, 2014)

Rescooped by Gilbert C FAURE from Cancer Vaccines Collection
Scoop.it!

Clinical deployment of antibodies for treatment of melanoma

Abstract

The concept of using immunotherapy to treat melanoma has existed for decades. The rationale comes from the knowledge that many patients with melanoma have endogenous immune responses against their tumor cells and clinically meaningful tumor regression can be achieved in a minority of patients using cytokines such as interleukin-2 and adoptive cellular therapy. In the last 5 years there has been a revolution in the clinical management of melanoma in large measure based on the development of antibodies that influence T cell regulatory pathways by overcoming checkpoint inhibition and providing co-stimulation, either of which results in significantly more effective immune-mediated tumor destruction. This review will describe the pre-clinical and clinical application of antagonistic antibodies targeting the T-cell checkpoints cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1), and agonistic antibodies targeting the costimulatory pathways OX40 and 4-1BB. Recent progress and opportunities for future investigation of combination antibody therapy will be described.


Via Krishan Maggon
Krishan Maggon 's curator insight, March 9, 2015 3:56 AM

Highlights

 

Melanoma treatment has been transformed using T-cell checkpoint antibodies.

Antibodies to CTLA-4 and PD-1 have had the largest impact on melanoma management.

Combination T-cell checkpoint therapy holds great promise for clinical development.

OX40 and 4-1BB are T cell costimulators with clinical potential in melanoma.

 

 

Rescooped by Gilbert C FAURE from Cancer Immunotherapy Review and Collection
Scoop.it!

Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies

Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies | Immunology and Biotherapies | Scoop.it
Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies http://t.co/4XGxIKdg1U #immunotherapy #lungcancer #awareness #LCAM @OxfordJournals

 

Abstract

Despite extensive investigation over the past three decades, cancer immunotherapy has produced limited success, with few agents achieving approval by the Food and Drug Administration and even the most effective helping only a minority of patients, primarily with melanoma or renal cancer. In recent years, immune checkpoints that maintain physiologic self-tolerance have been implicated in the down-regulation of anti-tumor immunity. Efforts to restore latent anti-tumor immunity have focused on antibody-based interventions targeting CTL antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) on T lymphocytes and its principal ligand (PD-L1) on tumor cells. Ipilimumab, an antibody targeting CTLA-4, appears to restore tumor immunity at the priming phase, whereas anti-PD-1/PD-L1 antibodies restore immune function in the tumor microenvironment. Although ipilimumab can produce durable long-term responses in patients with advanced melanoma, it is associated with significant immune-related toxicities. By contrast, antibodies targeting either PD-1 or PD-L1 have produced significant anti-tumor activity with considerably less toxicity. Activity was seen in patients with melanoma and renal cancer, as well as those with non-small-cell lung, bladder and head and neck cancers, tumors not previously felt to be sensitive to immunotherapy. The tolerability of PD-1-pathway blockers and their unique mechanism of action have made them ideal backbones for combination regimen development. Combination approaches involving cytotoxic chemotherapy, anti-angiogenic agents, alternative immune-checkpoint inhibitors, immunostimulatory cytokines and cancer vaccines are currently under clinical investigation. Current efforts focus on registration trials of single agents and combinations in various diseases and disease settings and identifying predictive biomarkers of response.

Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodiesTable 1.

Antibodies that target the PD-1 axis and are undergoing clinical investigation for cancer

 TargetAntibodyMolecular structureClinical development phaseTumor types in evaluationPD-1Nivolumab (BMS-936558)Fully human IgG4Phase IIIMelanoma, RCC, NSCLC, HNSCCPembrolizumab (MK-3475)Humanized IgG4Phase IIIMelanoma, NSCLCPidilizumab (CT-011)Humanized IgG1κPhase IIHEME, melanomaPD-L1BMS-936559Fully human IgG4Phase IAdvanced solid tumorsMPDL3280AFully human IgG1Phase IMelanoma, RCC, NSCLCPhase IIUROMEDI4736Fully human IgG1Phase IAdvanced solid tumorsPhase IIINSCLCMSB0010718CFully human IgG1Phase IAdvanced solid tumorsPhase IIMerkel cell carcinoma
Via Krishan Maggon
Krishan Maggon 's curator insight, November 15, 2014 1:38 AM

Review of the current status of active PD1/PDL1 projects                    

 

 

Int. Immunol. (2014)doi: 10.1093/intimm/dxu095                                     First published online: October 16, 2014

 

Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodiesGeorge K. Philips1 and Michael Atkins2

+Author Affiliations

1 Department of Medicine, Georgetown University Hospital, 3800 Reservoir Road NW, Washington DC 20007, USA2 Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Research Building-E501, 3970 Reservoir Road NW, Washington DC 20057, USACorrespondence to: G. K. Philips; E-mail: george.k.philips@gunet.georgetown.eduReceived September 4, 2014.Accepted October 3, 2014.
Krishan Maggon 's curator insight, November 15, 2014 2:06 AM

Review of the current status of active PD1/PDL1 projects                    

 

 

Int. Immunol. (2014)doi: 10.1093/intimm/dxu095                                     First published online: October 16, 2014

 

Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodiesGeorge K. Philips1 and Michael Atkins2

+Author Affiliations

1 Department of Medicine, Georgetown University Hospital, 3800 Reservoir Road NW, Washington DC 20007, USA2 Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Research Building-E501, 3970 Reservoir Road NW, Washington DC 20057, USACorrespondence to: G. K. Philips; E-mail: george.k.philips@gunet.georgetown.eduReceived September 4, 2014.Accepted October 3, 2014.