Network


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

Hotspot


Dive into the research topics where Kipp Weiskopf is active.

Publication


Featured researches published by Kipp Weiskopf.


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.


Nature | 2010

Asterless is a scaffold for the onset of centriole assembly

Nikola S. Dzhindzhev; Quan D. Yu; Kipp Weiskopf; George Tzolovsky; Inês Cunha-Ferreira; Maria Giovanna Riparbelli; Ana Rodrigues-Martins; Mónica Bettencourt-Dias; Giuliano Callaini; David M. Glover

Centrioles are found in the centrosome core and, as basal bodies, at the base of cilia and flagella. Centriole assembly and duplication is controlled by Polo-like-kinase 4 (Plk4): these processes fail if Plk4 is downregulated and are promoted by Plk4 overexpression. Here we show that the centriolar protein Asterless (Asl; human orthologue CEP152) provides a conserved molecular platform, the amino terminus of which interacts with the cryptic Polo box of Plk4 whereas the carboxy terminus interacts with the centriolar protein Sas-4 (CPAP in humans). Drosophila Asl and human CEP152 are required for the centrosomal loading of Plk4 in Drosophila and CPAP in human cells, respectively. Depletion of Asl or CEP152 caused failure of centrosome duplication; their overexpression led to de novo centriole formation in Drosophila eggs, duplication of free centrosomes in Drosophila embryos, and centrosome amplification in cultured Drosophila and human cells. Overexpression of a Plk4-binding-deficient mutant of Asl prevented centriole duplication in cultured cells and embryos. However, this mutant protein was able to promote microtubule organizing centre (MTOC) formation in both embryos and oocytes. Such MTOCs had pericentriolar material and the centriolar protein Sas-4, but no centrioles at their core. Formation of such acentriolar MTOCs could be phenocopied by overexpression of Sas-4 in oocytes or embryos. Our findings identify independent functions for Asl as a scaffold for Plk4 and Sas-4 that facilitates self-assembly and duplication of the centriole and organization of pericentriolar material.


Journal of Clinical Investigation | 2012

Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer

Holbrook Kohrt; Roch Houot; Kipp Weiskopf; Matthew J. Goldstein; Ferenc A. Scheeren; Debra K. Czerwinski; A. Dimitrios Colevas; Wen-Kai Weng; Michael F. Clarke; Robert W. Carlson; Frank E. Stockdale; Joseph A. Mollick; Lieping Chen; Ronald Levy

Trastuzumab, a monoclonal antibody targeting human epidermal growth factor receptor 2 (HER2; also known as HER-2/neu), is indicated for the treatment of women with either early stage or metastatic HER2(+) breast cancer. It kills tumor cells by several mechanisms, including antibody-dependent cellular cytotoxicity (ADCC). Strategies that enhance the activity of ADCC effectors, including NK cells, may improve the efficacy of trastuzumab. Here, we have shown that upon encountering trastuzumab-coated, HER2-overexpressing breast cancer cells, human NK cells become activated and express the costimulatory receptor CD137. CD137 activation, which was dependent on NK cell expression of the FcγRIII receptor, occurred both in vitro and in the peripheral blood of women with HER2-expressing breast cancer after trastuzumab treatment. Stimulation of trastuzumab-activated human NK cells with an agonistic mAb specific for CD137 killed breast cancer cells (including an intrinsically trastuzumab-resistant cell line) more efficiently both in vitro and in vivo in xenotransplant models of human breast cancer, including one using a human primary breast tumor. The enhanced cytotoxicity was restricted to antibody-coated tumor cells. This sequential antibody strategy, combining a tumor-targeting antibody with a second antibody that activates the host innate immune system, may improve the therapeutic effects of antibodies against breast cancer and other HER2-expressing tumors.


Blood | 2011

CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies

Holbrook Kohrt; Roch Houot; Matthew J. Goldstein; Kipp Weiskopf; Ash A. Alizadeh; Josh Brody; Antonia M. S. Müller; Russell Pachynski; Debra K. Czerwinski; Steven Coutre; Mark P. Chao; Lieping Chen; Thomas F. Tedder; Ronald Levy

Antibody-dependent cell-mediated cytotoxicity (ADCC), which is largely mediated by natural killer (NK) cells, is thought to play an important role in the efficacy of rituximab, an anti-CD20 monoclonal antibody (mAb) used to treat patients with B-cell lymphomas. CD137 is a costimulatory molecule expressed on a variety of immune cells after activation, including NK cells. In the present study, we show that an anti-CD137 agonistic mAb enhances the antilymphoma activity of rituximab by enhancing ADCC. Human NK cells up-regulate CD137 after encountering rituximab-coated tumor B cells, and subsequent stimulation of these NK cells with anti-CD137 mAb enhances rituximab-dependent cytotoxicity against the lymphoma cells. In a syngeneic murine lymphoma model and in a xenotransplanted human lymphoma model, sequential administration of anti-CD20 mAb followed by anti-CD137 mAb had potent antilymphoma activity in vivo. These results support a novel, sequential antibody approach against B-cell malignancies by targeting first the tumor and then the host immune system.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Anti-CD47 antibody–mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response

Diane Tseng; Jens-Peter Volkmer; Stephen B. Willingham; Humberto Contreras-Trujillo; John W. Fathman; Nathaniel B. Fernhoff; Jun Seita; Matthew A. Inlay; Kipp Weiskopf; Masanori Miyanishi; Irving L. Weissman

Mobilization of the T-cell response against cancer has the potential to achieve long-lasting cures. However, it is not known how to harness antigen-presenting cells optimally to achieve an effective antitumor T-cell response. In this study, we show that anti-CD47 antibody–mediated phagocytosis of cancer by macrophages can initiate an antitumor T-cell immune response. Using the ovalbumin model antigen system, anti-CD47 antibody–mediated phagocytosis of cancer cells by macrophages resulted in increased priming of OT-I T cells [cluster of differentiation 8-positive (CD8+)] but decreased priming of OT-II T cells (CD4+). The CD4+ T-cell response was characterized by a reduction in forkhead box P3-positive (Foxp3+) regulatory T cells. Macrophages following anti-CD47–mediated phagocytosis primed CD8+ T cells to exhibit cytotoxic function in vivo. This response protected animals from tumor challenge. We conclude that anti-CD47 antibody treatment not only enables macrophage phagocytosis of cancer but also can initiate an antitumor cytotoxic T-cell immune response.


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

Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma

Badreddin Edris; Kipp Weiskopf; Anne K. Volkmer; Jens-Peter Volkmer; Stephen B. Willingham; Humberto Contreras-Trujillo; Jie Liu; Ravindra Majeti; Robert B. West; Jonathan A. Fletcher; Andrew H. Beck; Irving L. Weissman; Matt van de Rijn

Antibodies against CD47, which block tumor cell CD47 interactions with macrophage signal regulatory protein-α, have been shown to decrease tumor size in hematological and epithelial tumor models by interfering with the protection from phagocytosis by macrophages that intact CD47 bestows upon tumor cells. Leiomyosarcoma (LMS) is a tumor of smooth muscle that can express varying levels of colony-stimulating factor-1 (CSF1), the expression of which correlates with the numbers of tumor-associated macrophages (TAMs) that are found in these tumors. We have previously shown that the presence of TAMs in LMS is associated with poor clinical outcome and the overall effect of TAMs in LMS therefore appears to be protumorigenic. However, the use of inhibitory antibodies against CD47 offers an opportunity to turn TAMs against LMS cells by allowing the phagocytic behavior of resident macrophages to predominate. Here we show that interference with CD47 increases phagocytosis of two human LMS cell lines, LMS04 and LMS05, in vitro. In addition, treatment of mice bearing subcutaneous LMS04 and LMS05 tumors with a novel, humanized anti-CD47 antibody resulted in significant reductions in tumor size. Mice bearing LMS04 tumors develop large numbers of lymph node and lung metastases. In a unique model for neoadjuvant treatment, mice were treated with anti-CD47 antibody starting 1 wk before resection of established primary tumors and subsequently showed a striking decrease in the size and number of metastases. These data suggest that treatment with anti-CD47 antibodies not only reduces primary tumor size but can also be used to inhibit the development of, or to eliminate, metastatic disease.


Journal of Clinical Investigation | 2014

Targeting CD137 enhances the efficacy of cetuximab

Holbrook Kohrt; A. Dimitrios Colevas; Roch Houot; Kipp Weiskopf; Matthew J. Goldstein; Peder Lund; A.M.S. Mueller; Idit Sagiv-Barfi; Aurélien Marabelle; Ruth Lira; Emily Troutner; Lori Richards; Amanda Rajapaska; Jonathan Hebb; Cariad Chester; Erin Waller; Anton Ostashko; Wen-Kai Weng; Lieping Chen; Debra K. Czerwinski; Yang-Xin Fu; John B. Sunwoo; Ronald Levy

Treatment with cetuximab, an EGFR-targeting IgG1 mAb, results in beneficial, yet limited, clinical improvement for patients with head and neck (HN) cancer as well as colorectal cancer (CRC) patients with WT KRAS tumors. Antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells contributes to the efficacy of cetuximab. The costimulatory molecule CD137 (4-1BB) is expressed following NK and memory T cell activation. We found that isolated human NK cells substantially increased expression of CD137 when exposed to cetuximab-coated, EGFR-expressing HN and CRC cell lines. Furthermore, activation of CD137 with an agonistic mAb enhanced NK cell degranulation and cytotoxicity. In multiple murine xenograft models, including EGFR-expressing cancer cells, HN cells, and KRAS-WT and KRAS-mutant CRC, combined cetuximab and anti-CD137 mAb administration was synergistic and led to complete tumor resolution and prolonged survival, which was dependent on the presence of NK cells. In patients receiving cetuximab, the level of CD137 on circulating and intratumoral NK cells was dependent on postcetuximab time and host FcyRIIIa polymorphism. Interestingly, the increase in CD137-expressing NK cells directly correlated to an increase in EGFR-specific CD8+ T cells. These results support development of a sequential antibody approach against EGFR-expressing malignancies that first targets the tumor and then the host immune system.


mAbs | 2015

Macrophages are critical effectors of antibody therapies for cancer.

Kipp Weiskopf; Irving L. Weissman

Macrophages are innate immune cells that derive from circulating monocytes, reside in all tissues, and participate in many states of pathology. Macrophages play a dichotomous role in cancer, where they promote tumor growth but also serve as critical immune effectors of therapeutic antibodies. Macrophages express all classes of Fcγ receptors, and they have immense potential to destroy tumors via the process of antibody-dependent phagocytosis. A number of studies have demonstrated that macrophage phagocytosis is a major mechanism of action of many antibodies approved to treat cancer. Consequently, a number of approaches to augment macrophage responses to therapeutic antibodies are under investigation, including the exploration of new targets and development of antibodies with enhanced functions. For example, the interaction of CD47 with signal-regulatory protein α (SIRPα) serves as a myeloid-specific immune checkpoint that limits the response of macrophages to antibody therapies, and CD47-blocking agents overcome this barrier to augment phagocytosis. The response of macrophages to antibody therapies can also be enhanced with engineered Fc variants, bispecific antibodies, or antibody-drug conjugates. Macrophages have demonstrated success as effectors of cancer immunotherapy, and further investigation will unlock their full potential for the benefit of patients.


Chemistry & Biology | 2012

A Nonpeptidic Cathepsin S Activity-Based Probe for Noninvasive Optical Imaging of Tumor-Associated Macrophages

Martijn Verdoes; Laura E. Edgington; Ferenc A. Scheeren; Melissa J. Leyva; Galia Blum; Kipp Weiskopf; Michael H. Bachmann; Jonathan A. Ellman; Matthew Bogyo

Macrophage infiltration into tumors has been correlated with poor clinical outcome in multiple cancer types. Therefore, tools to image tumor-associated macrophages could be valuable for diagnosis and prognosis of cancer. Herein, we describe the synthesis and characterization of a cathepsin S-directed, quenched activity-based probe (qABP), BMV083. This probe makes use of an optimized nonpeptidic scaffold leading to enhanced in vivo properties relative to previously reported peptide-based probes. In a syngeneic breast cancer model, BMV083 provides high tumor-specific fluorescence that can be visualized using noninvasive optical imaging methods. Furthermore, analysis of probe-labeled cells demonstrates that the probe primarily targets macrophages with an M2 phenotype. Thus, BMV083 is a potential valuable in vivo reporter for tumor-associated macrophages that could greatly facilitate the future studies of macrophage function in the process of tumorigenesis.


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.

Collaboration


Dive into the Kipp Weiskopf'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge