Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Praveen K. Bommareddy is active.

Publication


Featured researches published by Praveen K. Bommareddy.


American Journal of Clinical Dermatology | 2017

Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma.

Praveen K. Bommareddy; Anand Patel; Saamia Hossain; Howard L. Kaufman

Many mammalian viruses have properties that can be commandeered for the treatment of cancer. These characteristics include preferential infection and replication in tumor cells, the initiation of tumor cell lysis, and the induction of innate and adaptive anti-tumor immunity. Furthermore, viruses can be genetically engineered to reduce pathogenicity and increase immunogenicity resulting in minimally toxic therapeutic agents. Talimogene laherparepvec (T-VEC; Imlygic™), is a genetically modified herpes simplex virus, type 1, and is the first oncolytic virus therapy to be approved for the treatment of advanced melanoma by the US FDA. T-VEC is attenuated by the deletion of the herpes neurovirulence viral genes and enhanced for immunogenicity by the deletion of the viral ICP47 gene. Immunogenicity is further supported by expression of the human granulocyte–macrophage colony-stimulating factor (GM-CSF) gene, which helps promote the priming of T cell responses. T-VEC demonstrated significant improvement in durable response rate, objective response rate, and progression-free survival in a randomized phase III clinical trial for patients with advanced melanoma. This review will discuss the optimal selection of patients for such treatment and describe how therapy is optimally delivered. We will also discuss future directions for oncolytic virus immunotherapy, which will likely include combination T-VEC clinical trials, expansion of T-VEC to other types of non-melanoma skin cancers, and renewed efforts at oncolytic virus drug development with other viruses.


Frontiers in Oncology | 2017

Oncolytic Viruses—Natural and Genetically Engineered Cancer Immunotherapies

Sachin R. Jhawar; Aditya Thandoni; Praveen K. Bommareddy; Suemair Hassan; Frederick J. Kohlhapp; Sharad Goyal; Jason M. Schenkel; Ann W. Silk; Andrew Zloza

There has long been interest in innovating an approach by which tumor cells can be selectively and specifically targeted and destroyed. The discovery of viruses that lyse tumor cells, termed oncolytic viruses (OVs), has led to a revolution in the treatment of cancer. The potential of OVs to improve the therapeutic ratio is derived from their ability to preferentially infect and replicate in cancer cells while avoiding destruction of normal cells surrounding the tumor. Two main mechanisms exist through which these viruses are reported to improve outcomes: direct lysis of tumor cells and indirect augmentation of host anti-tumor immunity. With these factors in mind, viruses are chosen or modified to selectively target tumor cells, decrease pathogenicity to normal cells, decrease the antiviral immune response (to prevent viral clearance), and increase the antitumor immune response. While only one OV has been approved for the treatment of cancer in the United States, and only two other OVs have been approved worldwide, a wide spectrum of OVs are in various stages of preclinical development and in clinical trials. These viruses are being studied as alternatives and adjuncts to more traditional cancer therapies including surgical resection, chemotherapy, radiation, hormonal therapies, targeted therapies, and other immunotherapies. Here, we review the natural characteristics and genetically engineered modifications that enhance the effectiveness of OVs for the treatment of cancer.


Cancer Research | 2017

Abstract CT026: Phase 1b study of intratumoral Coxsackievirus A21 (CVA21) and systemicpembrolizumab inadvanced melanoma patients: Interim results of the CAPRA clinical trial

Ann W. Silk; Howard L. Kaufman; Nashat Y. Gabrail; Janice M. Mehnert; Jennifer Bryan; Jacqueline Norrell; Daniel Medina; Praveen K. Bommareddy; Darren R. Shafren; Mark Grose; Andrew Zloza

Background: Coxsackievirus A21 (CVA21) is a novel bio-selected oncolytic, immunotherapeutic agent. Intratumoral (i.t.) CVA21 injection can induce selective tumor-cell infection, immune-cell infiltration, IFN-γ response gene up-regulation, increased PD-L1 expression, tumor cell lysis and systemic anti-tumor immune responses. Preclinical studies in an immune-competent mouse model of melanoma have revealed that combinations of i.t. CVA21 and anti-PD-1 blockade mediate significantly greater antitumor activity compared to use of either agent alone. A clinical trial evaluating combination CVA21 and pembrolizumab in patients with melanoma was initiated and preliminary data on a pre-established futility endpoint are presented here. Materials and Methods: This is a single-arm, multi-institutional open-label phase Ib clinical trial of i.t. CVA21 and i.v. pembrolizumab for treated or untreated unresectable Stage IIIC-IVM1c melanoma. Subjects with injectable disease receive up to 3 x 10 8 TCID 50 CVA21 i.t. on Days 1, 3, 5, 8, and then every 3 weeks for up to 19 injections. Subjects also receive pembrolizumab (2mg/kg) i.v. every 3 weeks starting on Day 8. The primary endpoint is safety/tolerability by incidence of dose-limiting toxicity. Secondary endpoints include best ORR by immune-related response criteria, progression-free survival, overall survival, quality of life, changes in melanoma-specific T cells, PD-L1 expression and Th1/Th2 gene expression profiles. The protocol included a futility analysis after the first 12 patients. Results : To date, 14 subjects have started on protocol therapy. Overall, the adverse events have been low-grade constitutional symptoms related to CVA21 and expected pembrolizumab-related side effects. No DLT’s have been reported. Currently, 11 patients are evaluable for investigator response assessment, not including 2 subjects who have not yet reached their first assessment and 1 subject who left the study early due to an unrelated adverse event. Among the evaluable subjects, the ORR was 73% (8/11). The DCR (CR+PR+SD) is currently 91% (10/11). In subjects with stage IVM1c disease, the ORR and the DCR is 100% (5/5). The study has met its primary statistical futility endpoint of achieving ≥2 confirmed objective responses (CR or PR) in the first 12 patients enrolled. Currently, the median time to response is 1.6 months. One of the 8 responders displayed early pseudo-progression and later developed a partial response. Conclusions: At a pre-specified futility analysis, combination CVA21 and pembrolizumab appears to be well-tolerated. Early tumor monitoring has identified encouraging reductions in a number of injected and non-injected lesions. Based on these initial results, the sample size has now been expanded to enroll up to 50 patients. Combination therapy of CVA21 and pembrolizumab may represent a new approach for the treatment of patients with injectable advanced melanoma. Citation Format: Ann W. Silk, Howard Kaufman, Nashat Gabrail, Janice Mehnert, Jennifer Bryan, Jacqueline Norrell, Daniel Medina, Praveen Bommareddy, Darren Shafren, Mark Grose, Andrew Zloza. Phase 1b study of intratumoral Coxsackievirus A21 ( C V A 21) and systemic p emb r olizumab in a dvanced melanoma patients: Interim results of the CAPRA clinical trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT026. doi:10.1158/1538-7445.AM2017-CT026


Nature Reviews Immunology | 2018

Integrating oncolytic viruses in combination cancer immunotherapy

Praveen K. Bommareddy; Megha Shettigar; Howard L. Kaufman

Oncolytic viruses can be usefully integrated into tumour immunotherapies, as they target multiple steps within the cancer–immunity cycle. Oncolytic viruses directly lyse tumour cells, leading to the release of soluble antigens, danger signals and type I interferons, which drive antitumour immunity. In addition, some oncolytic viruses can be engineered to express therapeutic genes or can functionally alter tumour-associated endothelial cells, further enhancing T cell recruitment into immune-excluded or immune-deserted tumour microenvironments. Oncolytic viruses can also utilize established tumours as an in situ source of neoantigen vaccination through cross-presentation, resulting in regression of distant, uninfected tumours. These features make oncolytic viruses attractive agents for combination strategies to optimize cancer immunotherapy.Oncolytic viruses can target multiple steps in the cancer–immunity cycle and can be engineered to express therapeutic genes and, as a result, can be usefully integrated in combination tumour immunotherapies. Here, the authors discuss features of oncolytic viruses that make them appealing agents for combination approaches in cancer immunotherapy.


Journal of Surgical Oncology | 2018

Metastasectomy following incomplete response to high-dose interleukin-2

Tasha Hughes; Joseph Broucek; Gail Iodice; Praveen K. Bommareddy; Sanjib Basu; Howard L. Kaufman

To evaluate our experience with metastasectomy following partial response or stable disease after treatment with high‐dose interleukin‐2 (HD IL‐2).


Journal of Clinical Investigation | 2018

Unleashing the therapeutic potential of oncolytic viruses

Praveen K. Bommareddy; Howard L. Kaufman

Oncolytic viruses (OVs) are a versatile new class of therapeutic agents based on native or genetically modified viruses that selectively replicate in tumor cells and can express therapeutic transgenes designed to target cells within the tumor microenvironment and/or host immunity. To date, however, confirmation of the underlying mechanism of action and an understanding of innate and acquired drug resistance for most OVs have been limited. In this issue of the JCI, Zamarin et al. report a comprehensive analysis of an oncolytic Newcastle disease virus (NDV) using both murine melanoma tumor models and human tumor explants to explore how the virus promotes tumor eradication and details of the mechanisms involved. These findings have implications for the optimization of oncolytic immunotherapy, at least that based on NDV, and further confirm that specific combinatorial approaches are promising for clinical development.


Oncogene | 2018

Novel bone morphogenetic protein receptor inhibitor JL5 suppresses tumor cell survival signaling and induces regression of human lung cancer

Jenna Newman; David J. Augeri; Rachel NeMoyer; Jyoti Malhotra; Elaine Langenfeld; Charles B. Chesson; Natalie S. Dobias; Michael J. Lee; Saeed Tarabichi; Sachin R. Jhawar; Praveen K. Bommareddy; Sh’Rae Marshall; Evita T. Sadimin; John E. Kerrigan; Michael J. Goedken; Christine Minerowicz; Salma K. Jabbour; Shengguo Li; Mary O. Carayannopolous; Andrew Zloza; John Langenfeld

BMP receptor inhibitors induce death of cancer cells through the downregulation of antiapoptotic proteins XIAP, pTAK1, and Id1-Id3. However, the current most potent BMP receptor inhibitor, DMH2, does not downregulate BMP signaling in vivo because of metabolic instability and poor pharmacokinetics. Here we identified the site of metabolic instability of DMH2 and designed a novel BMP receptor inhibitor, JL5. We show that JL5 has a greater volume of distribution and suppresses the expression of Id1 and pTak1 in tumor xenografts. Moreover, we demonstrate JL5-induced tumor cell death and tumor regression in xenograft mouse models without immune cells and humanized with adoptively transferred human immune cells. In humanized mice, JL5 additionally induces the infiltration of immune cells within the tumor microenvironment. Our studies show that the BMP signaling pathway is targetable in vivo and BMP receptor inhibitors can be developed as a therapeutic to treat cancer patients.


Nature Reviews Immunology | 2018

Author Correction: Integrating oncolytic viruses in combination cancer immunotherapy

Praveen K. Bommareddy; Megha Shettigar; Howard L. Kaufman

In the initially published version of this article online in advance of print, a reference (Ajina, A. & Maher, J. Prospects for combined use of oncolytic viruses and CAR T-cells. J. Immunother. Cancer 5, 90 (2017)) was omitted in error from the following sentence: “The ability of oncolytic viruses to increase the expression of MHC class I molecules by cancer cells is also predicted to enhance ACT with TILs or TCR-engineered and chimeric antigen receptor (CAR) T cells that target tumour-specific antigens”. This has been corrected in the HTML and PDF versions of the manuscript.


Future Oncology | 2017

Avelumab and other recent advances in Merkel cell carcinoma

Praveen K. Bommareddy; Howard L. Kaufman

Merkel cell carcinoma (MCC) is a rare but aggressive form of skin cancer that occurs in the elderly, is associated with UV radiation and immunosuppression. Initial treatment consists of wide excision with adjuvant radiation. Although the tumor is sensitive to chemotherapy, long-term survival is unusual and there had been no US FDA-approved drugs prior to 2017. The recognition that MCC is associated with the Merkel cell polyomavirus occurs more commonly in immune-compromised patients and tumors express PD-L1 suggested testing immunotherapy. A study of an anti-PD-L1 antibody, avelumab, in chemotherapy-refractory MCC demonstrated a response rate of 31.8% resulting in FDA approval in March 2017 and EMA in September 2017. This review will discuss the disease, role of avelumab and other emerging treatment strategies for MCC.


Cancer Journal | 2017

Intratumoral Approaches for the Treatment of Melanoma

Praveen K. Bommareddy; Ann W. Silk; Howard L. Kaufman

Collaboration


Dive into the Praveen K. Bommareddy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles B. Chesson

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge