Network


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

Hotspot


Dive into the research topics where Lee P. Richman is active.

Publication


Featured researches published by Lee P. Richman.


Journal of Clinical Investigation | 2013

Human Treg responses allow sustained recombinant adeno-associated virus–mediated transgene expression

Christian Mueller; Jeffrey D. Chulay; Bruce C. Trapnell; Margaret Humphries; Brenna Carey; Robert A. Sandhaus; Noel G. McElvaney; Louis M. Messina; Qiushi Tang; Farshid N. Rouhani; Martha Campbell-Thompson; Ann Dongtao Fu; Anthony T. Yachnis; David R. Knop; Guo-jie Ye; Mark L. Brantly; Roberto Calcedo; Suryanarayan Somanathan; Lee P. Richman; Robert H. Vonderheide; Maigan A. Hulme; Todd M. Brusko; James M. Wilson; Terence R. Flotte

Recombinant adeno-associated virus (rAAV) vectors have shown promise for the treatment of several diseases; however, immune-mediated elimination of transduced cells has been suggested to limit and account for a loss of efficacy. To determine whether rAAV vector expression can persist long term, we administered rAAV vectors expressing normal, M-type α-1 antitrypsin (M-AAT) to AAT-deficient subjects at various doses by multiple i.m. injections. M-specific AAT expression was observed in all subjects in a dose-dependent manner and was sustained for more than 1 year in the absence of immune suppression. Muscle biopsies at 1 year had sustained AAT expression and a reduction of inflammatory cells compared with 3 month biopsies. Deep sequencing of the TCR Vβ region from muscle biopsies demonstrated a limited number of T cell clones that emerged at 3 months after vector administration and persisted for 1 year. In situ immunophenotyping revealed a substantial Treg population in muscle biopsy samples containing AAT-expressing myofibers. Approximately 10% of all T cells in muscle were natural Tregs, which were activated in response to AAV capsid. These results suggest that i.m. delivery of rAAV type 1-AAT (rAAV1-AAT) induces a T regulatory response that allows ongoing transgene expression and indicates that immunomodulatory treatments may not be necessary for rAAV-mediated gene therapy.


PLOS ONE | 2016

Antifibrotic Effects of the Dual CCR2/CCR5 Antagonist Cenicriviroc in Animal Models of Liver and Kidney Fibrosis.

Eric Lefebvre; Graeme Moyle; Ran Reshef; Lee P. Richman; Melanie Thompson; Feng Hong; Hsin-l Chou; Taishi Hashiguchi; Craig F. Plato; Dominic Poulin; Toni L. Richards; Hiroyuki Yoneyama; Helen E. Jenkins; Grushenka Wolfgang; Scott L. Friedman

Background & Aims Interactions between C-C chemokine receptor types 2 (CCR2) and 5 (CCR5) and their ligands, including CCL2 and CCL5, mediate fibrogenesis by promoting monocyte/macrophage recruitment and tissue infiltration, as well as hepatic stellate cell activation. Cenicriviroc (CVC) is an oral, dual CCR2/CCR5 antagonist with nanomolar potency against both receptors. CVC’s anti-inflammatory and antifibrotic effects were evaluated in a range of preclinical models of inflammation and fibrosis. Methods Monocyte/macrophage recruitment was assessed in vivo in a mouse model of thioglycollate-induced peritonitis. CCL2-induced chemotaxis was evaluated ex vivo on mouse monocytes. CVC’s antifibrotic effects were evaluated in a thioacetamide-induced rat model of liver fibrosis and mouse models of diet-induced non-alcoholic steatohepatitis (NASH) and renal fibrosis. Study assessments included body and liver/kidney weight, liver function test, liver/kidney morphology and collagen deposition, fibrogenic gene and protein expression, and pharmacokinetic analyses. Results CVC significantly reduced monocyte/macrophage recruitment in vivo at doses ≥20 mg/kg/day (p < 0.05). At these doses, CVC showed antifibrotic effects, with significant reductions in collagen deposition (p < 0.05), and collagen type 1 protein and mRNA expression across the three animal models of fibrosis. In the NASH model, CVC significantly reduced the non-alcoholic fatty liver disease activity score (p < 0.05 vs. controls). CVC treatment had no notable effect on body or liver/kidney weight. Conclusions CVC displayed potent anti-inflammatory and antifibrotic activity in a range of animal fibrosis models, supporting human testing for fibrotic diseases. Further experimental studies are needed to clarify the underlying mechanisms of CVC’s antifibrotic effects. A Phase 2b study in adults with NASH and liver fibrosis is fully enrolled (CENTAUR Study 652-2-203; NCT02217475).


Cancer immunology research | 2014

Role of Crosslinking for Agonistic CD40 Monoclonal Antibodies as Immune Therapy of Cancer

Lee P. Richman; Robert H. Vonderheide

In a comprehensive analysis of the agonistic activity of anti-human CD40 mAb CP-870,893, Richman and Vonderheide show that Fc-crosslinking of CP-870,893 is not required. In contrast, the therapeutic potency is more dependent on the CD40 epitopes recognized. Agonists of the TNF superfamily of receptors hold promise as novel therapy for cancer. Recent data on agonistic antimurine TNF receptors (TNFR) such as CD40 suggest that the specific engagement of Fc receptor (FcR) is required for optimal antitumor effects, prompting calls to engineer antihuman CD40 and other TNFR monoclonal antibodies (mAb) accordingly. CP-870,893 is a fully human anti-CD40 mAb, selected in part because it is an immunoglobulin G2 (IgG2), which is presumed to have poor reactivity with FcR; however, CP-870,893 has been evaluated in multiple clinical trials with beneficial activity in patients with melanoma, pancreatic, and other cancers. Here, we confirmed that the activity of antimurine CD40 mAb was dependent on FcγRIIB engagement, was decreased significantly in FcγRIIB−/− mice, and upon Fc-crosslinking antimouse CD40 mAb enhanced the activation of antigen-presenting cells. In contrast, the CP-870,893-mediated activation of human B cells was not enhanced with anti-IgG crosslinking nor abrogated when used as an F(ab)′2 reagent. Crosslinking of CP-870,893 using the CD32-expressing K562 cells yielded an Fc-dependent modest increase in the expression of some activation markers relative to that of the soluble CP-870,893 mAb. Classic Fc-dependent functions such as antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CMC) were minimal for CP-870,893 as compared with the IgG1 anti-CD20 mAb rituximab, which mediated both ADCC and CMC in parallel assays. Antimouse CD40 mAb competed for the CD40 ligand binding site, but CP-870,893 did not. Thus, Fc crosslinking is not an essential requirement for agonistic antihuman CD40 mAbs, in which potency is more dependent on the CD40 epitope recognized and the strength of the signal achieved. Cancer Immunol Res; 2(1); 19–26. ©2013 AACR.


Journal of Clinical Oncology | 2015

High Graft CD8 Cell Dose Predicts Improved Survival and Enables Better Donor Selection in Allogeneic Stem-Cell Transplantation With Reduced-Intensity Conditioning

Ran Reshef; Austin P. Huffman; Amy Gao; Marlise R. Luskin; Noelle V. Frey; Saar Gill; Elizabeth O. Hexner; Taku Kambayashi; Alison W. Loren; Selina M. Luger; James K. Mangan; Sunita D. Nasta; Lee P. Richman; Mary Sell; Edward A. Stadtmauer; Robert H. Vonderheide; Rosemarie Mick; David L. Porter

PURPOSE To characterize the impact of graft T-cell composition on outcomes of reduced-intensity conditioned (RIC) allogeneic hematopoietic stem-cell transplantation (alloHSCT) in adults with hematologic malignancies. PATIENTS AND METHODS We evaluated associations between graft T-cell doses and outcomes in 200 patients who underwent RIC alloHSCT with a peripheral blood stem-cell graft. We then studied 21 alloHSCT donors to identify predictors of optimal graft T-cell content. RESULTS Higher CD8 cell doses were associated with a lower risk for relapse (adjusted hazard ratio [aHR], 0.43; P = .009) and improved relapse-free survival (aHR, 0.50; P = .006) and overall survival (aHR, 0.57; P = .04) without a significant increase in graft-versus-host disease or nonrelapse mortality. A cutoff level of 0.72 × 10(8) CD8 cells per kilogram optimally segregated patients receiving CD8(hi) and CD8(lo) grafts with differing overall survival (P = .007). Donor age inversely correlated with graft CD8 dose. Consequently, older donors were unlikely to provide a CD8(hi) graft, whereas approximately half of younger donors provided CD8(hi) grafts. Compared with recipients of older sibling donor grafts (consistently containing CD8(lo) doses), survival was significantly better for recipients of younger unrelated donor grafts with CD8(hi) doses (P = .03), but not for recipients of younger unrelated donor CD8(lo) grafts (P = .28). In addition, graft CD8 content could be predicted by measuring the proportion of CD8 cells in a screening blood sample from stem-cell donors. CONCLUSION Higher graft CD8 dose, which was restricted to young donors, predicted better survival in patients undergoing RIC alloHSCT.


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


Blood | 2015

NKG2D expression by CD8 + T-cells contributes to GVHD and GVT effects in a murine model of allogeneic HSCT

Mobin Karimi; Jerrod L. Bryson; Lee P. Richman; Andrew D. Fesnak; Theresa M. Leichner; Atsushi Satake; Robert H. Vonderheide; David H. Raulet; Ran Reshef; Taku Kambayashi

In allogeneic hematopoietic stem cell transplantation (HSCT), controlling graft-versus-host disease (GVHD) while maintaining graft-versus-tumor (GVT) responses is of critical importance. Using a mouse model of allogeneic HSCT, we hereby demonstrate that NKG2D expression by CD8(+) T cells plays a major role in mediating GVHD and GVT effects by promoting the survival and cytotoxic function of CD8(+) T cells. The expression of NKG2D ligands was not induced persistently on normal tissues of allogeneic HSCT-recipient mice treated with anti-NKG2D antibody, suggesting that transient NKG2D blockade might be sufficient to attenuate GVHD and allow CD8(+) T cells to regain their GVT function. Indeed, short-term treatment with anti-NKG2D antibody restored GVT effects while maintaining an attenuated GVHD state. NKG2D expression was also detected on CD8(+) T cells from allogeneic HSCT patients and trended to be higher in those with active GVHD. Together, these data support a novel role for NKG2D expression by CD8(+) T cells during allogeneic HSCT, which could be potentially therapeutically exploited to separate GVHD from GVT effects.


Blood | 2017

Clinical and immunologic impact of CCR5 blockade in graft-versus-host disease prophylaxis

Austin P. Huffman; Lee P. Richman; Lisa Crisalli; Ximi K. Wang; James A. Hoxie; Rosemarie Mick; Stephen G. Emerson; Yi Zhang; Robert H. Vonderheide; David L. Porter; Ran Reshef

Graft-versus-host disease (GVHD) is a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Lymphocyte trafficking via chemokine receptors such as CCR5 plays a critical role in alloreactive responses, and previous data suggest that CCR5 blockade with maraviroc results in a low incidence of visceral GVHD. However, the full scope of clinical and immunologic effects of CCR5 blockade in HSCT has not been described. We compared a cohort of patients enrolled on a trial of reduced-intensity allo-HSCT with standard GVHD prophylaxis plus maraviroc to a contemporary control cohort receiving standard GVHD prophylaxis alone. Maraviroc treatment was associated with a lower incidence of acute GVHD without increased risk of disease relapse, as well as reduced levels of gut-specific markers. At day 30, maraviroc treatment increased CCR5 expression on T cells and dampened T-cell activation in peripheral blood without impairing early immune reconstitution or increasing risk for infections. Patients who developed acute GVHD despite maraviroc prophylaxis showed increased T-cell activation, naive T-cell skewing, and elevated serum CXCL9 and CXCL10 levels. Collectively, these data suggest that maraviroc effectively protects against GVHD by modulating alloreactive donor T-cell responses, and that CXCR3 signaling may be an important resistance mechanism to CCR5 blockade in GVHD.


OncoImmunology | 2014

Anti-human CD40 monoclonal antibody therapy is potent without FcR crosslinking

Lee P. Richman; Robert H. Vonderheide

Antibody agonists targeting tumor necrosis factor (TNF) superfamily receptors, including CD40, are being tested therapeutically as anticancer agents. Studies in mice have shown that anti-CD40 monoclonal antibody (mAb) requires Fc-receptor (FcR) engagement to activate antitumor immunity. In contrast, we have reported that clinically active anti-human CD40 mAb CP-870,893 does not require FcR crosslinking, a finding with translational implications.


Immunity | 2018

Tumor Cell-Intrinsic Factors Underlie Heterogeneity of Immune Cell Infiltration and Response to Immunotherapy

Jinyang Li; Katelyn T. Byrne; Fangxue Yan; Taiji Yamazoe; Zeyu Chen; Timour Baslan; Lee P. Richman; Jeffrey H. Lin; Yu H. Sun; Andrew J. Rech; David Balli; Ceire A. Hay; Yogev Sela; Allyson J. Merrell; Shannon M. Liudahl; Naomi Gordon; Robert J. Norgard; Salina Yuan; Sixiang Yu; Timothy Chao; Shuai Ye; T.S. Karin Eisinger-Mathason; Robert B. Faryabi; John W. Tobias; Scott W. Lowe; Lisa M. Coussens; E. John Wherry; Robert H. Vonderheide; Ben Z. Stanger

Summary The biological and functional heterogeneity between tumors—both across and within cancer types—poses a challenge for immunotherapy. To understand the factors underlying tumor immune heterogeneity and immunotherapy sensitivity, we established a library of congenic tumor cell clones from an autochthonous mouse model of pancreatic adenocarcinoma. These clones generated tumors that recapitulated T cell‐inflamed and non‐T‐cell‐inflamed tumor microenvironments upon implantation in immunocompetent mice, with distinct patterns of infiltration by immune cell subsets. Co‐injecting tumor cell clones revealed the non‐T‐cell‐inflamed phenotype is dominant and that both quantitative and qualitative features of intratumoral CD8+ T cells determine response to therapy. Transcriptomic and epigenetic analyses revealed tumor‐cell‐intrinsic production of the chemokine CXCL1 as a determinant of the non‐T‐cell‐inflamed microenvironment, and ablation of CXCL1 promoted T cell infiltration and sensitivity to a combination immunotherapy regimen. Thus, tumor cell‐intrinsic factors shape the tumor immune microenvironment and influence the outcome of immunotherapy. Graphical Abstract Figure. No caption available. HighlightsGenerated a library of congenic pancreatic cancer cell clones derived from KPC miceEach tumor elicited unique immune infiltration correlating with therapeutic responseTumors lacking T cells exhibit different epigenetic and transcriptomic statusCXCL1 was increased in therapy‐resistant tumors that lacked T cell infiltration &NA; Using a library of pancreatic cancer cell clones, Li et al. identify heterogeneous and multifactorial pathways regulating tumor‐cell‐intrinsic mechanisms that dictate the immune microenvironment and thereby responses to immunotherapy. This tumor clone library provides a tool for identifying new targets responsible for thwarting responses to immunotherapy in resistant tumors.

Collaboration


Dive into the Lee P. Richman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ran Reshef

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

David L. Porter

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rosemarie Mick

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Austin P. Huffman

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Noelle V. Frey

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Alison W. Loren

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James K. Mangan

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge