Network


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

Hotspot


Dive into the research topics where David L. Bajor is active.

Publication


Featured researches published by David L. Bajor.


Journal of Clinical Oncology | 2007

Clinical Activity and Immune Modulation in Cancer Patients Treated With CP-870,893, a Novel CD40 Agonist Monoclonal Antibody

Robert H. Vonderheide; Keith T. Flaherty; Magi Khalil; Molly S. Stumacher; David L. Bajor; Natalie A. Hutnick; Patricia Sullivan; J. Joseph Mahany; Maryann Gallagher; Amy Kramer; Stephanie Green; Peter J. O'Dwyer; Kelli L. Running; Richard D. Huhn; Scott Antonia

PURPOSE The cell-surface molecule CD40 activates antigen-presenting cells and enhances immune responses. CD40 is also expressed by solid tumors, but its engagement results in apoptosis. CP-870,893, a fully human and selective CD40 agonist monoclonal antibody (mAb), was tested for safety in a phase I dose-escalation study. PATIENTS AND METHODS Patients with advanced solid tumors received single doses of CP-870,893 intravenously. The primary objective was to determine safety and the maximum-tolerated dose (MTD). Secondary objectives included assessment of immune modulation and tumor response. RESULTS Twenty-nine patients received CP-870,893 in doses from 0.01 to 0.3 mg/kg. Dose-limiting toxicity was observed in two of seven patients at the 0.3 mg/kg dose level (venous thromboembolism and grade 3 headache). MTD was estimated as 0.2 mg/kg. The most common adverse event was cytokine release syndrome (grade 1 to 2) which included chills, rigors, and fever. Transient laboratory abnormalities affecting lymphocytes, monocytes, platelets, D-dimer and liver function tests were observed 24 to 48 hours after infusion. Four patients with melanoma (14% of all patients and 27% of melanoma patients) had objective partial responses at restaging (day 43). CP-870,893 infusion resulted in transient depletion of CD19+ B cells in blood (93% depletion at the MTD for < 1 week). Among B cells remaining in blood, we found a dose-related upregulation of costimulatory molecules after treatment. CONCLUSION The CD40 agonist mAb CP-870,893 was well tolerated and biologically active, and was associated with antitumor activity. Further studies of repeated doses of CP-870,893 alone and in combination with other antineoplastic agents are warranted.


Cancer immunology research | 2015

Induction of T cell immunity overcomes complete resistance to PD-1 and CTLA-4 blockade and improves survival in pancreatic carcinoma

Rafael Winograd; Katelyn T. Byrne; Rebecca A. Evans; Pamela M. Odorizzi; Anders R.L. Meyer; David L. Bajor; Cynthia Clendenin; Ben Z. Stanger; Emma E. Furth; E. John Wherry; Robert H. Vonderheide

Winograd and colleagues used an engineered KPC mouse model of pancreatic ductal adenocarcinoma (PDA) reflecting that of human PDA to show that baseline refractoriness to checkpoint inhibitors can be rescued by priming a T-cell response with αCD40 plus chemotherapy with gemcitabine and nab-paclitaxel. Disabling the function of immune checkpoint molecules can unlock T-cell immunity against cancer, yet despite remarkable clinical success with monoclonal antibodies (mAb) that block PD-1 or CTLA-4, resistance remains common and essentially unexplained. To date, pancreatic carcinoma is fully refractory to these antibodies. Here, using a genetically engineered mouse model of pancreatic ductal adenocarcinoma in which spontaneous immunity is minimal, we found that PD-L1 is prominent in the tumor microenvironment, a phenotype confirmed in patients; however, tumor PD-L1 was found to be independent of IFNγ in this model. Tumor T cells expressed PD-1 as prominently as T cells from chronically infected mice, but treatment with αPD-1 mAbs, with or without αCTLA-4 mAbs, failed in well-established tumors, recapitulating clinical results. Agonist αCD40 mAbs with chemotherapy induced T-cell immunity and reversed the complete resistance of pancreatic tumors to αPD-1 and αCTLA-4. The combination of αCD40/chemotherapy plus αPD-1 and/or αCTLA-4 induced regression of subcutaneous tumors, improved overall survival, and conferred curative protection from multiple tumor rechallenges, consistent with immune memory not otherwise achievable. Combinatorial treatment nearly doubled survival of mice with spontaneous pancreatic cancers, although no cures were observed. Our findings suggest that in pancreatic carcinoma, a nonimmunogenic tumor, baseline refractoriness to checkpoint inhibitors can be rescued by the priming of a T-cell response with αCD40/chemotherapy. Cancer Immunol Res; 3(4); 399–411. ©2015 AACR.


Cancer Immunology, Immunotherapy | 2013

CD40 immunotherapy for pancreatic cancer

Robert H. Vonderheide; David L. Bajor; Rafael Winograd; Rebecca A. Evans; Lauren J. Bayne; Gregory L. Beatty

Pancreatic ductal adenocarcinoma (PDA) is a highly aggressive and lethal cancer which is poorly responsive to standard therapies. Although the PDA tumor microenvironment is considered especially immunosuppressive, recent data mostly from genetically engineered and other mouse models of the disease suggest that novel immunotherapeutic approaches hold promise. Here, we describe both laboratory and clinical efforts to target the CD40 pathway for immunotherapy in PDA. Findings suggest that CD40 agonists can mediate both T-cell-dependent and T-cell-independent immune mechanisms of tumor regression in mice and patients. T-cell-independent mechanisms are associated with macrophage activation and the destruction of PDA tumor stroma, supporting the concept that immune modulation of the tumor microenvironment represents a useful approach in cancer immunotherapy.


Nature Communications | 2016

Metastatic progression is associated with dynamic changes in the local microenvironment

Nicole M. Aiello; David L. Bajor; Robert J. Norgard; Amine Sahmoud; Neha Bhagwat; Minh N. Pham; Toby C. Cornish; Christine A. Iacobuzio-Donahue; Robert H. Vonderheide; Ben Z. Stanger

Most cancer-associated deaths result from metastasis. However, it remains unknown whether the size, microenvironment or other features of a metastatic lesion dictate its behaviour or determine the efficacy of chemotherapy in the adjuvant (micrometastatic) setting. Here we delineate the natural history of metastasis in an autochthonous model of pancreatic ductal adenocarcinoma (PDAC), using lineage tracing to examine the evolution of disseminated cancer cells and their associated microenvironment. With increasing size, lesions shift from mesenchymal to epithelial histology, become hypovascular and accumulate a desmoplastic stroma, ultimately recapitulating the primary tumours from which they arose. Moreover, treatment with gemcitabine and nab-paclitaxel significantly reduces the overall number of metastases by inducing cell death in lesions of all sizes, challenging the paradigm that PDAC stroma imposes a critical barrier to drug delivery. These results illuminate the cellular dynamics of metastatic progression and suggest that adjuvant chemotherapy affords a survival benefit by directly targeting micrometastases.


Cancer immunology research | 2014

Immune activation and a 9-year ongoing complete remission following CD40 antibody therapy and metastasectomy in a patient with metastatic melanoma.

David L. Bajor; Xiaowei Xu; Drew A. Torigian; Rosemarie Mick; Laura R. Garcia; Lee P. Richman; Cindy Desmarais; Katherine L. Nathanson; Lynn M. Schuchter; Michael Kalos; Robert H. Vonderheide

Bajor and colleagues used next-generation sequencing of TCRs in the tumor and blood to identify the emergence and persistence of a de novo T-cell repertoire following agonistic CD40 therapy, highlighting the potential of immune agonists in therapy and TCR deep sequencing in immune assessment. Direct immune activation via agonistic mAbs is a potentially complementary approach to therapeutic blockade of inhibitory immune receptors in cancer. Here, we provide genetic analysis of the immunologic consequences associated with the use of an agonistic CD40 mAb in a patient with metastatic melanoma who responded, underwent a single metastasectomy, and then achieved a complete remission ongoing for more than 9 years after starting therapy. Tumor microenvironment after immunotherapy was associated with proinflammatory modulations and emergence of a de novo T-cell repertoire as detected by next-generation sequencing of T-cell receptors (TCR) in the tumor and blood. The de novo T-cell repertoire identified in the posttreatment metastasectomy sample was also present—and in some cases expanded—in the circulation years after completion of therapy. Comprehensive study of this “exceptional responder” highlights the emerging potential of direct immune agonists in the next wave of cancer immunotherapies and a potential role for TCR deep sequencing in cancer immune assessment. Cancer Immunol Res; 2(11); 1051–8. ©2014 AACR.


Cancer Research | 2015

Abstract CT137: Combination of agonistic CD40 monoclonal antibody CP-870,893 and anti-CTLA-4 antibody tremelimumab in patients with metastatic melanoma

David L. Bajor; Rosemarie Mick; Matthew J. Riese; Lee P. Richman; Xiaowei Xu; Drew A. Torigian; Erietta Stelekati; Martha Sweeney; Brendan Sullivan; Lynn M. Schuchter; Ravi K. Amaravadi; E. John Wherry; Robert H. Vonderheide

Purpose: Combining immunostimulatory monoclonal (mAb) and checkpoint inhibition may improve response rates and overall survival in patients with metastatic melanoma. CP-870,893 is an agonistic fully human IgG2 mAb targeting the immune stimulatory molecule CD40 (αCD40). Tremelimumab (treme) is a fully human IgG2 mAb targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA4), a negative co-stimulatory molecule found on T cells. Methods: Patients with metastatic melanoma were enrolled on a single-arm open-label phase I study. αCD40 and treme were dosed i.v. every 3 weeks and 12 weeks, respectively. Dose escalation followed a 3 + 3 strategy with alternating increases of αCD40 and treme in each cohort. Outcomes were scored using the Common Terminology Criteria for Adverse Events v3.0 and RECIST 1.0. Flow cytometric analysis of baseline blood samples was compared to samples obtained at least 3 weeks after the last dose of αCD40 and treme. Results: Toxicity: Twenty-four patients were enrolled and treated at 4 different dose levels. Two patients were replaced per protocol due to rapid, symptomatic progression of disease; these patients were not evaluated for response but were included in toxicity analysis. Dose-limiting toxicities (DLT), considered to be likely due to treatment, were colitis (n = 1) and hypophysitis (n = 1) and uveitis (n = 1). Cytokine release syndrome (CRS), grade 1-2, occurring within 24 hours of CP administration, was the most common treatment-related toxicity occurring in 19 (79.2%) patients. CRS symptoms were controlled with standard supportive care. All episodes of CRS resolved within 24 hours of αCD40 administration. The estimated maximum tolerated dose was 0.2 mg/kg of αCD40 and 10 mg/kg of treme. Outcomes: Overall objective response rate (ORR) was 27.3% (best response): two patients (9.1%) had complete responses to treatment and four (18.2%) patients had partial responses. The median follow-up was 22 months with a median progression free survival of 2.5 months and a median overall survival (OS) of 26.1 months. Correlative studies: Flow cytometric analysis of peripheral blood lymphocytes revealed changes in CD8+ T cell phenotypes. Double positive granzyme B+ and Ki-67+ cells increased from 0.56 ± 0.17% (mean ± standard error) for all CD8+ T cells at baseline to 1.01 ± 0.23% post-treatment (p = 0.03). CD8+ T cells expressing both programmed cell death 1 and eomesodermin were also higher after treatment, increasing from 7.7 ± 1.1% to 10.4 ±1.8% (p = 0.04) of all CD8+ T cells. Conclusion: Combination therapy with αCD40 and treme was well-tolerated in patients with metastatic melanoma, with rates of CRS and other toxicity similar to previously reported rates for αCD40 or treme treatments alone. Overall ORR was 27.3%, with median OS of 26.1 months. Given the tolerability, antitumor activity, and biomarker evidence of immune activation, expanded study of this combination should be pursued. Citation Format: David L. Bajor, Rosemarie Mick, Matthew J. Riese, Lee P. Richman, Xiaowei Xu, Drew A. Torigian, Erietta Stelekati, Martha Sweeney, Brendan Sullivan, Lynn M. Schuchter, Ravi Amaravadi, E. John Wherry, Robert H. Vonderheide. Combination of agonistic CD40 monoclonal antibody CP-870,893 and anti-CTLA-4 antibody tremelimumab in patients with metastatic melanoma. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT137. doi:10.1158/1538-7445.AM2015-CT137


Journal of Immunology | 2016

CSF-1R-Dependent Lethal Hepatotoxicity When Agonistic CD40 Antibody Is Given before but Not after Chemotherapy.

Katelyn T. Byrne; Nathan H. Leisenring; David L. Bajor; Robert H. Vonderheide

Cancer immunotherapies are increasingly effective in the clinic, especially immune checkpoint blockade delivered to patients who have T cell–infiltrated tumors. Agonistic CD40 mAb promotes stromal degradation and, in combination with chemotherapy, drives T cell infiltration and de novo responses against tumors, rendering resistant tumors susceptible to current immunotherapies. Partnering anti-CD40 with different treatments is an attractive approach for the next phase of cancer immunotherapies, with a number of clinical trials using anti-CD40 combinations ongoing, but the optimal therapeutic regimens with anti-CD40 are not well understood. Pancreatic ductal adenocarcinoma (PDA) is classically resistant to immunotherapy and lacks baseline T cell infiltration. In this study, we used a tumor cell line derived from a genetically engineered mouse model of PDA to investigate alterations in the sequence of anti-CD40 and chemotherapy as an approach to enhance pharmacological delivery of chemotherapy. Unexpectedly, despite our previous studies showing anti-CD40 treatment after chemotherapy is safe in both mice and patients with PDA, we report in this article that anti-CD40 administration <3 d in advance of chemotherapy is lethal in more than half of treated C57BL/6 mice. Anti-CD40 treatment 2 or 3 d before chemotherapy resulted in significantly increased populations of both activated myeloid cells and macrophages and lethal hepatotoxicity. Liver damage was fully abrogated when macrophage activation was blocked using anti–CSF-1R mAb. These studies highlight the dual nature of CD40 in activating both macrophages and T cell responses, and the need for preclinical investigation of optimal anti-CD40 treatment regimens for safe design of clinical trials.


Clinical Cancer Research | 2012

Rehabilitation for Oncogene Addiction: Role of Immunity in Cellular Sobriety

David L. Bajor; Robert H. Vonderheide

Clinical responses to oncogene inhibitors result from direct effects on cell-intrinsic growth signals and disruption of downstream messages that produce a protumor immunosuppressive microenvironment. Combining oncogene-targeted and immunomodulatory therapies may result in synergistic effects, producing increased response rates and longer periods of tumor control than can be achieved with either class alone. Clin Cancer Res; 18(5); 1192–4. ©2012 AACR.


Journal of Immunotherapy | 2013

Cracking the stone: combination vaccination and CTLA-4 blockade in pancreatic cancer.

David L. Bajor; Robert H. Vonderheide

Despite an extensive immunosuppressive microenvironment that encircles pancreatic ductal adenocarcinoma (PDA) even at the earliest stages of the disease, pancreatic cancer cells may be paradoxically sensitive to T-cell attack, because a mechanism of “immune privilege” limits the tumor’s encounter with adaptive immune cells. “Darwinian” immune pressure to drive selection of immunoresistant clones appears to be minimal.1 This biology may provide an important clinical opportunity for strategies that combine active vaccination and blockade of immunosuppressive negative checkpoints relevant in PDA. The clinical findings reported by Le et al2 support this notion and offer hope that such combination strategies may be added to the armamentarium for treatment of this recalcitrant tumor. In this study, patients with advanced, refractory PDA received the anti-CTLA-4 mAb ipilimumab alone or in combination with an allogeneic tumor cell genetically engineered to express granulocyte-macrophage colony-stimulating factor (GM-CSF) (GVAX). Both ipilimumab3 and GVAX4–6 have been previously explored in patients with pancreatic cancer, but never together in this clinical setting. Ipilimumab in combination with gemcitabine chemotherapy is associated with minimal clinical activity, and although GVAX vaccination in the adjuvant setting in patients with resected pancreatic cancer has shown promise,4,5 GVAX alone for patients with metastatic pancreatic cancer has been much less active.6 Because both GVAX and ipilimumab have an acceptable safety profile (ipilimumab being FDA approved for melanoma), Le and colleagues combined the two approaches to test for clinical and immunologic synergy in a randomized study. The results are striking, provocative, and carry major implications for the design of immunotherapy in pancreatic and other cancers. Although the study was not powered to demonstrate statistically significant differences in overall survival (OS), Le and colleagues nevertheless observed that 27% of patients in the combination treatment arm were alive at 1 year compared with 7% of patients in the ipilimumab-only arm. Moreover, longer survival was associated with an increase in peak induction of mesothelin-specific T cells and the maintenance of a larger mesothelin-specific T-cell repertoire after completion of treatment. Patients who did not live to the median overall survival time point of 4.7 months did not maintain mesothelin-specific T-cell responses. Importantly, the addition of GVAX to ipilimumab did not increase toxicity. Despite ipilimumab dosing of 10mg/kg, which is >3 times higher than the current dose approved in melanoma, treatments were tolerated similarly in both groups, with 20% of patients experiencing grades 3 to 4 immune-related adverse events compared with a rate of approximately 15% seen in other studies in melanoma at the 3mg/kg dose level. It should be noted that no objective tumor responses as calculated with RECIST criteria were observed in either arm. Mechanistically, GVAX is thought to prime an immune response to PDA, whereas ipilimumab inhibits both direct suppressive effects of CTLA-4 ligation on effector T cells and possibly depletes inhibitory regulatory T cells,7,8 which are common and functional in PDA. It is interesting to note that Le and colleagues show that the combination of GVAX does not substantially enhance the rate of immune-related adverse events seen with ipilimumab alone, and there was no autoimmune toxicity attributable to the mesothelin-specific T cells such as pleuritis or pericarditis. In preclinical studies that date back more than a decade, CTLA-4 blockade and GVAX demonstrated antitumor efficacy that is dependent on both CD4 and CD8 T cells and capable of producing immunity to repeated tumor challenge.9 When ipilimumab and GVAX were combined in previous, early-phase trials for the treatment of patients with chemotherapy-naive,


OncoImmunology | 2018

Long-term outcomes of a phase I study of agonist CD40 antibody and CTLA-4 blockade in patients with metastatic melanoma

David L. Bajor; Rosemarie Mick; Matthew J. Riese; Alex C. Huang; Brendan Sullivan; Lee P. Richman; Drew A. Torigian; Sangeeth M. George; Erietta Stelekati; Fang Chen; Joseph Melenhorst; Simon F. Lacey; Xiaowei Xu; E. John Wherry; Tara C. Gangadhar; Ravi K. Amaravadi; Lynn M. Schuchter; Robert H. Vonderheide

ABSTRACT We report long-term clinical outcomes and immune responses observed from a phase 1 trial of agonist CD40 monoclonal antibody (mAb) and blocking CTLA-4 mAb in patients with metastatic melanoma. Twenty-four patients previously untreated with checkpoint blockade were enrolled. The agonistic CD40 mAb CP-870,893 and the CTLA-4 blocking mAb tremelimumab were dosed concomitantly every 3 weeks and 12 weeks, respectively, across four dose combinations. Two patients developed dose-limiting grade 3 immune-mediated colitis that led to the definition of the maximum tolerated dose (MTD). Other immune-mediated toxicity included uveitis (n = 1), hypophysitis (n = 1), hypothyroidism (n = 2), and grade 3 cytokine release syndrome (CRS) (n = 1). The estimated MTD was 0.2 mg/kg of CP-870,893 and 10 mg/kg of tremelimumab. In 22 evaluable patients, the objective response rate (ORR) was 27.3%: two patients (9.1%) had complete responses (CR) and four (18.2%) patients had partial responses (PR). With a median follow-up of 45 months, the median progression-free survival (PFS) was 3.2 months (95% CI, 1.3–5.1 months) and median overall survival (OS) was 23.6 months (95% CI, 11.7–35.5 months). Nine patients are long-term survivors (> 3 years), 8 of whom subsequently received other therapy including PD-1 mAb, surgery, or radiation therapy. Elevated baseline soluble CD25 was associated with shorter OS. Immunologically, treatment was associated with evidence of T cell activation and increased tumor T cell infiltration that was accomplished without therapeutic PD-1/PD-L1 blockade. These results suggest opportunities for immune activation and cancer immunotherapy beyond PD-1.

Collaboration


Dive into the David L. Bajor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ben Z. Stanger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rosemarie Mick

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Drew A. Torigian

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

E. John Wherry

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Lee P. Richman

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Lynn M. Schuchter

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Xiaowei Xu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Brendan Sullivan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Erietta Stelekati

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge