Network


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

Hotspot


Dive into the research topics where Jens Peter Volkmer is active.

Publication


Featured researches published by Jens Peter Volkmer.


Nature Biotechnology | 2011

An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells

Chad Tang; Andrew S. Lee; Jens Peter Volkmer; Debashis Sahoo; Divya Nag; Adriane R. Mosley; Matthew A. Inlay; Reza Ardehali; Shawn L. Chavez; Renee A. Reijo Pera; B. Behr; Joseph C. Wu; Irving L. Weissman; Micha Drukker

An important risk in the clinical application of human pluripotent stem cells (hPSCs), including human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), is teratoma formation by residual undifferentiated cells. We raised a monoclonal antibody against hESCs, designated anti–stage-specific embryonic antigen (SSEA)-5, which binds a previously unidentified antigen highly and specifically expressed on hPSCs—the H type-1 glycan. Separation based on SSEA-5 expression through fluorescence-activated cell sorting (FACS) greatly reduced teratoma-formation potential of heterogeneously differentiated cultures. To ensure complete removal of teratoma-forming cells, we identified additional pluripotency surface markers (PSMs) exhibiting a large dynamic expression range during differentiation: CD9, CD30, CD50, CD90 and CD200. Immunohistochemistry studies of human fetal tissues and bioinformatics analysis of a microarray database revealed that concurrent expression of these markers is both common and specific to hPSCs. Immunodepletion with antibodies against SSEA-5 and two additional PSMs completely removed teratoma-formation potential from incompletely differentiated hESC cultures.


Science Translational Medicine | 2010

Calreticulin Is the Dominant Pro-Phagocytic Signal on Multiple Human Cancers and Is Counterbalanced by CD47

Mark P. Chao; Siddhartha Jaiswal; Rachel Weissman-Tsukamoto; Ash A. Alizadeh; Andrew J. Gentles; Jens Peter Volkmer; Kipp Weiskopf; Stephen B. Willingham; Tal Raveh; Christopher Y. Park; Ravindra Majeti; Irving L. Weissman

Calreticulin-induced phagocytosis of cancer cells can be counterbalanced by CD47 expression. Eat Up! Immune cells constantly patrol the body on a search and destroy campaign against foreign invaders. Designed to detect differential molecular signals, cells of the immune system can distinguish healthy from infected tissue by the types of proteins produced: Infected cells, for example, often produce unfamiliar proteins, which then activate innate immune cells to “eat” (phagocytose) the infected ones. Cancer cells also carry aberrant cargo such as unfamiliar proteins or normal proteins at abnormal levels, yet these cells frequently escape immune attack because they express a “don’t eat me” signal, the cell surface protein CD47. Blocking this signal on a cancer cell makes them targets for phagocytosis, but surprisingly does not do the same for normal cells that express CD47. Chao et al. have now identified calreticulin as the “eat me” signal on cancer cells that leads to phagocytosis when the counterbalancing “don’t eat me” signal CD47 is blocked. Calreticulin is a pro-phagocytic molecule that is highly expressed on the surface of several types of human cancer cells, including acute myeloid and lymphoblastic leukemias, chronic myeloid leukemia, non-Hodgkin’s lymphoma, bladder cancer, glioblastoma, and ovarian cancer. However, calreticulin is expressed only at very low levels on normal cells. Chao et al. found a correlation between calreticulin and CD47 expression levels on cancer cells and showed that blocking the interaction between calreticulin and its ligand prevented phagocytosis initiated by blocking the “don’t eat me” signal CD47. Moreover, high calreticulin expression on cancer cells was a poor prognostic indicator in human patients with neuroblastoma, bladder cancer, and non-Hodgkin’s lymphoma. Therefore, a balance between calreticulin and CD47 expression in cancer cells may be a double-edged sword: In the absence of a CD47 blocker, this equilibrium may support tumor cell survival, but when CD47 function is inhibited, the presence of calreticulin tells immune cells to “eat up!” This information provides a key insight for the therapeutic development of CD47-inhibitory agents. Under normal physiological conditions, cellular homeostasis is partly regulated by a balance of pro- and anti-phagocytic signals. CD47, which prevents cancer cell phagocytosis by the innate immune system, is highly expressed on several human cancers including acute myeloid leukemia, non-Hodgkin’s lymphoma, and bladder cancer. Blocking CD47 with a monoclonal antibody results in phagocytosis of cancer cells and leads to in vivo tumor elimination, yet normal cells remain mostly unaffected. Thus, we postulated that cancer cells must also display a potent pro-phagocytic signal. Here, we identified calreticulin as a pro-phagocytic signal that was highly expressed on the surface of several human cancers, but was minimally expressed on most normal cells. Increased CD47 expression correlated with high amounts of calreticulin on cancer cells and was necessary for protection from calreticulin-mediated phagocytosis. Blocking the interaction of target cell calreticulin with its receptor, low-density lipoprotein receptor–related protein, on phagocytic cells prevented anti-CD47 antibody–mediated phagocytosis. Furthermore, increased calreticulin expression was an adverse prognostic factor in diverse tumors including neuroblastoma, bladder cancer, and non-Hodgkin’s lymphoma. These findings identify calreticulin as the dominant pro-phagocytic signal on several human cancers, provide an explanation for the selective targeting of tumor cells by anti-CD47 antibody, and highlight the balance between pro- and anti-phagocytic signals in the immune evasion of cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Three differentiation states risk-stratify bladder cancer into distinct subtypes

Jens Peter Volkmer; Debashis Sahoo; Robert K. Chin; Philip Levy Ho; Chad Tang; Antonina V. Kurtova; Stephen B. Willingham; Senthil Pazhanisamy; Humberto Contreras-Trujillo; Theresa A. Storm; Yair Lotan; Andrew H. Beck; Benjamin I. Chung; Ash A. Alizadeh; Guilherme Godoy; Seth P. Lerner; Matt van de Rijn; Linda D. Shortliffe; Irving L. Weissman; Keith Syson Chan

Current clinical judgment in bladder cancer (BC) relies primarily on pathological stage and grade. We investigated whether a molecular classification of tumor cell differentiation, based on a developmental biology approach, can provide additional prognostic information. Exploiting large preexisting gene-expression databases, we developed a biologically supervised computational model to predict markers that correspond with BC differentiation. To provide mechanistic insight, we assessed relative tumorigenicity and differentiation potential via xenotransplantation. We then correlated the prognostic utility of the identified markers to outcomes within gene expression and formalin-fixed paraffin-embedded (FFPE) tissue datasets. Our data indicate that BC can be subclassified into three subtypes, on the basis of their differentiation states: basal, intermediate, and differentiated, where only the most primitive tumor cell subpopulation within each subtype is capable of generating xenograft tumors and recapitulating downstream populations. We found that keratin 14 (KRT14) marks the most primitive differentiation state that precedes KRT5 and KRT20 expression. Furthermore, KRT14 expression is consistently associated with worse prognosis in both univariate and multivariate analyses. We identify here three distinct BC subtypes on the basis of their differentiation states, each harboring a unique tumor-initiating population.


PLOS ONE | 2015

Pre-Clinical Development of a Humanized Anti-CD47 Antibody with Anti-Cancer Therapeutic Potential

Jie Liu; Lijuan Wang; Feifei Zhao; Serena Tseng; Cyndhavi Narayanan; Lei Shura; Stephen B. Willingham; Maureen Howard; Susan S. Prohaska; Jens Peter Volkmer; Mark P. Chao; Irving L. Weissman; Ravindra Majeti

CD47 is a widely expressed cell surface protein that functions as a regulator of phagocytosis mediated by cells of the innate immune system, such as macrophages and dendritic cells. CD47 serves as the ligand for a receptor on these innate immune cells, SIRP-alpha, which in turn delivers an inhibitory signal for phagocytosis. We previously found increased expression of CD47 on primary human acute myeloid leukemia (AML) stem cells, and demonstrated that blocking monoclonal antibodies directed against CD47 enabled the phagocytosis and elimination of AML, non-Hodgkin’s lymphoma (NHL), and many solid tumors in xenograft models. Here, we report the development of a humanized anti-CD47 antibody with potent efficacy and favorable toxicokinetic properties as a candidate therapeutic. A novel monoclonal anti-human CD47 antibody, 5F9, was generated, and antibody humanization was carried out by grafting its complementarity determining regions (CDRs) onto a human IgG4 format. The resulting humanized 5F9 antibody (Hu5F9-G4) bound monomeric human CD47 with an 8 nM affinity. Hu5F9-G4 induced potent macrophage-mediated phagocytosis of primary human AML cells in vitro and completely eradicated human AML in vivo, leading to long-term disease-free survival of patient-derived xenografts. Moreover, Hu5F9-G4 synergized with rituximab to eliminate NHL engraftment and cure xenografted mice. Finally, toxicokinetic studies in non-human primates showed that Hu5F9-G4 could be safely administered intravenously at doses able to achieve potentially therapeutic serum levels. Thus, Hu5F9-G4 is actively being developed for and has been entered into clinical trials in patients with AML and solid tumors (ClinicalTrials.gov identifier: NCT02216409).


Science Translational Medicine | 2014

Endoscopic molecular imaging of human bladder cancer using a CD47 antibody

Ying Pan; Jens Peter Volkmer; Kathleen E. Mach; Robert V. Rouse; Jen Jane Liu; Debashis Sahoo; Timothy Chang; Thomas J. Metzner; Lei Kang; Matt van de Rijn; Eila C. Skinner; Sanjiv S. Gambhir; Irving L. Weissman; Joseph C. Liao

Fluorescently labeled CD47 antibody is an endoscopic molecular imaging agent to diagnose human bladder cancer. Lighting Up Bladder Cancer Lesions Molecular imaging of bladder cancer could greatly improve on current methods of diagnosis, which rely on white light–based imaging that looks for superficial tissue changes, such as color and texture. To this end, Pan et al. targeted the cancer-specific marker CD47 that is present on the surface of solid tumors, including in the bladder. The authors attached a brightly fluorescent tag, called a quantum dot, to a CD47 antibody. The fluorescent antibody was instilled into human bladders with muscle and nonmuscle invasive cancer that had been recently removed from patients. Overall, 119 bladder regions were analyzed using the fluorescent antibody and blue light cystoscopy. The authors reported a sensitivity of 82.9% and a specificity of 90.5%. They further correctly identified five of six carcinoma in situ lesions—a diagnostic challenge for white light imaging. Because this approach only requires topical administration of a fluorescent antibody and the use of already available clinical tools, it is hoped that this molecular imaging approach to diagnosing various bladder cancers will translate after further optimization. A combination of optical imaging technologies with cancer-specific molecular imaging agents is a potentially powerful strategy to improve cancer detection and enable image-guided surgery. Bladder cancer is primarily managed endoscopically by white light cystoscopy with suboptimal diagnostic accuracy. Emerging optical imaging technologies hold great potential for improved diagnostic accuracy but lack imaging agents for molecular specificity. Using fluorescently labeled CD47 antibody (anti-CD47) as molecular imaging agent, we demonstrated consistent identification of bladder cancer with clinical grade fluorescence imaging systems, confocal endomicroscopy, and blue light cystoscopy in fresh surgically removed human bladders. With blue light cystoscopy, the sensitivity and specificity for CD47-targeted imaging were 82.9 and 90.5%, respectively. We detected variants of bladder cancers, which are diagnostic challenges, including carcinoma in situ, residual carcinoma in tumor resection bed, recurrent carcinoma following prior intravesical immunotherapy with Bacillus Calmette-Guérin (BCG), and excluded cancer from benign but suspicious-appearing mucosa. CD47-targeted molecular imaging could improve diagnosis and resection thoroughness for bladder cancer.


Journal of Clinical Investigation | 2016

CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer.

Kipp Weiskopf; Nadine S. Jahchan; Peter J. Schnorr; Sandra Cristea; Aaron M. Ring; Roy L. Maute; Anne K. Volkmer; Jens Peter Volkmer; Jie Liu; Jing Shan Lim; Dian Yang; Garrett Seitz; Thuyen Nguyen; Di Wu; Kevin M. Jude; Heather Guerston; Francesca Trapani; Julie George; John T. Poirier; Eric E. Gardner; Linde A. Miles; Elisa de Stanchina; Shane Lofgren; Hannes Vogel; Monte M. Winslow; Caroline Dive; Roman K. Thomas; Charles M. Rudin; Matt Van De Rijn; Ravindra Majeti

Small-cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer with limited treatment options. CD47 is a cell-surface molecule that promotes immune evasion by engaging signal-regulatory protein alpha (SIRPα), which serves as an inhibitory receptor on macrophages. Here, we found that CD47 is highly expressed on the surface of human SCLC cells; therefore, we investigated CD47-blocking immunotherapies as a potential approach for SCLC treatment. Disruption of the interaction of CD47 with SIRPα using anti-CD47 antibodies induced macrophage-mediated phagocytosis of human SCLC patient cells in culture. In a murine model, administration of CD47-blocking antibodies or targeted inactivation of the Cd47 gene markedly inhibited SCLC tumor growth. Furthermore, using comprehensive antibody arrays, we identified several possible therapeutic targets on the surface of SCLC cells. Antibodies to these targets, including CD56/neural cell adhesion molecule (NCAM), promoted phagocytosis in human SCLC cell lines that was enhanced when combined with CD47-blocking therapies. In light of recent clinical trials for CD47-blocking therapies in cancer treatment, these findings identify disruption of the CD47/SIRPα axis as a potential immunotherapeutic strategy for SCLC. This approach could enable personalized immunotherapeutic regimens in patients with SCLC and other cancers.


Nature Cell Biology | 2014

A cell-intrinsic role for TLR2–MYD88 in intestinal and breast epithelia and oncogenesis

Ferenc A. Scheeren; Angera H. Kuo; Linda J. van Weele; Shang Cai; Iris Glykofridis; Shaheen S. Sikandar; Maider Zabala; Dalong Qian; Jessica Lam; Darius M. Johnston; Jens Peter Volkmer; Debashis Sahoo; Matt van de Rijn; Frederick M. Dirbas; George Somlo; Tomer Kalisky; Michael E. Rothenberg; Stephen R. Quake; Michael F. Clarke

It has been postulated that there is a link between inflammation and cancer. Here we describe a role for cell-intrinsic toll-like receptor-2 (TLR2; which is involved in inflammatory response) signalling in normal intestinal and mammary epithelial cells and oncogenesis. The downstream effectors of TLR2 are expressed by normal intestinal and mammary epithelia, including the stem/progenitor cells. Deletion of MYD88 or TLR2 in the intestinal epithelium markedly reduces DSS-induced colitis regeneration and spontaneous tumour development in mice. Limiting dilution transplantations of breast epithelial cells devoid of TLR2 or MYD88 revealed a significant decrease in mammary repopulating unit frequency compared with the control. Inhibition of TLR2, its co-receptor CD14, or its downstream targets MYD88 and IRAK1 inhibits growth of human breast cancers in vitro and in vivo. These results suggest that inhibitors of the TLR2 pathway merit investigation as possible therapeutic and chemoprevention agents.


Science Translational Medicine | 2016

Hematopoietic stem cell transplantation in immunocompetent hosts without radiation or chemotherapy

Akanksha Chhabra; Aaron M. Ring; Kipp Weiskopf; Peter J. Schnorr; Sydney R. Gordon; Alan C. Le; Hye-Sook Kwon; Nan Guo Ring; Jens Peter Volkmer; Po Yi Ho; Serena Tseng; Irving L. Weissman; Judith A. Shizuru

Combining anti–c-Kit antibodies with CD47 blockade depletes endogenous hematopoietic stem cells and enables bone marrow transplantation without radiation or chemotherapeutics. Make way for stem cells Current conditioning regimens to prepare the host bone marrow for transplantation of donor hematopoietic stem cells (HSCs) can have toxic effects on the patient. As an alternative to chemotherapy or radiation, Chhabra et al. describe a new immunotherapy strategy that clears out host HSCs from the bone marrow in preparation for a transplant. Blockade of the surface antigen CD47 allows phagocytic myeloid cells to engulf host HSCs displaced by antibody targeting, effectively depleting HSCs from the bone marrow of immunocompetent mice. This led to improved engraftment of donor HSCs, with fewer toxic effects for the mice receiving the transplant. Hematopoietic stem cell (HSC) transplantation can cure diverse diseases of the blood system, including hematologic malignancies, anemias, and autoimmune disorders. However, patients must undergo toxic conditioning regimens that use chemotherapy and/or radiation to eliminate host HSCs and enable donor HSC engraftment. Previous studies have shown that anti–c-Kit monoclonal antibodies deplete HSCs from bone marrow niches, allowing donor HSC engraftment in immunodeficient mice. We show that host HSC clearance is dependent on Fc-mediated antibody effector functions, and enhancing effector activity through blockade of CD47, a myeloid-specific immune checkpoint, extends anti–c-Kit conditioning to fully immunocompetent mice. The combined treatment leads to elimination of >99% of host HSCs and robust multilineage blood reconstitution after HSC transplantation. This targeted conditioning regimen that uses only biologic agents has the potential to transform the practice of HSC transplantation and enable its use in a wider spectrum of patients.


Science Translational Medicine | 2017

Disrupting the CD47-SIRPα anti-phagocytic axis by a humanized anti-CD47 antibody is an efficacious treatment for malignant pediatric brain tumors

Sharareh Gholamin; Siddhartha Mitra; Abdullah H. Feroze; Jie Liu; Suzana Assad Kahn; Michael Zhang; Rogelio Esparza; Chase Richard; Vijay Ramaswamy; Marc Remke; Anne K. Volkmer; Stephen B. Willingham; Anitha Ponnuswami; Aaron McCarty; Patricia Lovelace; Theresa A. Storm; Simone Schubert; Gregor Hutter; Cyndhavi Narayanan; Pauline Chu; Eric Raabe; Griffith R. Harsh; Michael D. Taylor; Michelle Monje; Yoon Jae Cho; Ravi Majeti; Jens Peter Volkmer; Paul G. Fisher; Gerald A. Grant; Gary K. Steinberg

Anti-CD47 antibody is effective for treating malignant pediatric brain tumors without detectable toxicity in patient-derived xenograft models. Brain tumors, meet macrophages A protein called CD47 is often expressed on the surface of tumor cells, where it serves as a “don’t eat me” signal that blocks macrophages from attacking the tumor. To overcome this signal and allow the macrophages to “eat” tumor cells, Gholamin et al. engineered a humanized antibody that blocks CD47 signaling. The researchers tested the efficacy of this antibody in patient-derived xenograft models of a variety of pediatric brain tumors. The treatment was successful at inhibiting CD47, killing tumor cells, and prolonging the animals’ survival, all without toxic effects on normal tissues. Morbidity and mortality associated with pediatric malignant primary brain tumors remain high in the absence of effective therapies. Macrophage-mediated phagocytosis of tumor cells via blockade of the anti-phagocytic CD47-SIRPα interaction using anti-CD47 antibodies has shown promise in preclinical xenografts of various human malignancies. We demonstrate the effect of a humanized anti-CD47 antibody, Hu5F9-G4, on five aggressive and etiologically distinct pediatric brain tumors: group 3 medulloblastoma (primary and metastatic), atypical teratoid rhabdoid tumor, primitive neuroectodermal tumor, pediatric glioblastoma, and diffuse intrinsic pontine glioma. Hu5F9-G4 demonstrated therapeutic efficacy in vitro and in vivo in patient-derived orthotopic xenograft models. Intraventricular administration of Hu5F9-G4 further enhanced its activity against disseminated medulloblastoma leptomeningeal disease. Notably, Hu5F9-G4 showed minimal activity against normal human neural cells in vitro and in vivo, a phenomenon reiterated in an immunocompetent allograft glioma model. Thus, Hu5F9-G4 is a potentially safe and effective therapeutic agent for managing multiple pediatric central nervous system malignancies.


Experimental Hematology | 2015

HSC transplantation in an immunocompetent host without radiation or chemotherapy

Akanksha Chhabra; Aaron M. Ring; Kipp Weiskopf; Peter J. Schnorr; Sydney R. Gordon; Alan C. Le; Hye-Sook Kwon; Nan Guo; Jens Peter Volkmer; Po Yi Ho; Serena Tseng; Irving L. Weissman; Judith A. Shizuru

Collaboration


Dive into the Jens Peter Volkmer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Debashis Sahoo

University of California

View shared research outputs
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
Researchain Logo
Decentralizing Knowledge