Network


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

Hotspot


Dive into the research topics where Robbie G. Majzner is active.

Publication


Featured researches published by Robbie G. Majzner.


Nature Medicine | 2017

CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy

Terry J. Fry; Nirali N. Shah; Rimas J. Orentas; Maryalice Stetler-Stevenson; Constance Yuan; Sneha Ramakrishna; Pamela L. Wolters; Staci Martin; Cindy Delbrook; Bonnie Yates; Haneen Shalabi; Thomas J. Fountaine; Jack F. Shern; Robbie G. Majzner; David F. Stroncek; Marianna Sabatino; Yang Feng; Dimiter S. Dimitrov; Ling Zhang; Sang Nguyen; Haiying Qin; Boro Dropulic; Daniel W. Lee; Crystal L. Mackall

Chimeric antigen receptor (CAR) T cells targeting CD19 mediate potent effects in relapsed and/or refractory pre–B cell acute lymphoblastic leukemia (B-ALL), but antigen loss is a frequent cause of resistance to CD19-targeted immunotherapy. CD22 is also expressed in most cases of B-ALL and is usually retained following CD19 loss. We report results from a phase 1 trial testing a new CD22-targeted CAR (CD22-CAR) in 21 children and adults, including 17 who were previously treated with CD19-directed immunotherapy. Dose-dependent antileukemic activity was observed, with complete remission obtained in 73% (11/15) of patients receiving ≥1 × 106 CD22-CAR T cells per kg body weight, including 5 of 5 patients with CD19dim or CD19− B-ALL. Median remission duration was 6 months. Relapses were associated with diminished CD22 site density that likely permitted CD22+ cell escape from killing by CD22-CAR T cells. These results are the first to establish the clinical activity of a CD22-CAR in B-ALL, including leukemia resistant to anti-CD19 immunotherapy, demonstrating potency against B-ALL comparable to that of CD19-CAR at biologically active doses. Our results also highlight the critical role played by antigen density in regulating CAR function.Chimeric antigen receptor (CAR) T-cells targeting CD19 mediate potent effects in relapsed/refractory pre-B cell acute lymphoblastic leukemia (B-ALL) but antigen loss is a frequent cause of resistance to CD19-targeted immunotherapy. CD22 is also expressed on most B-ALL and usually retained following CD19 loss. We report results from a phase I trial testing a novel CD22-CAR in twenty-one children and adults, including 17 previously treated with CD19-directed immunotherapy. Dose dependent anti-leukemic activity was observed with complete remission in 73% (11/15) of patients receiving ≥ 1 × 106 CD22-CART cells/kg, including 5/5 patients with CD19dim/neg B-ALL. Median remission duration was 6 months. Relapses were associated with diminished CD22 site density that likely permitted escape from killing by CD22-CART cells. These results are the first to eastablish the clinical activity of a CD22-CAR in pre-B cell ALL, including in leukemia resistant to anti-CD19 immunotherapy, demonstrating comparable potency to CD19-CART at biologically active doses in B-ALL. They also highlight the critical role played by antigen density in regulating CAR function. (Funded by NCI Intramural Research Program)


British Journal of Haematology | 2012

T cell depletion utilizing CD34+ stem cell selection and CD3+ addback from unrelated adult donors in paediatric allogeneic stem cell transplantation recipients

Mark B. Geyer; Angela Ricci; Judith S. Jacobson; Robbie G. Majzner; Deirdre Duffy; Carmella van de Ven; Janet Ayello; Monica Bhatia; James Garvin; Diane George; Prakash Satwani; Lauren Harrison; Erin Morris; Mildred Semidei-Pomales; Joseph E. Schwartz; Bachir Alobeid; Lee Ann Baxter-Lowe; Mitchell S. Cairo

CD34‐selected haploidentical and unrelated donor allogeneic stem cell transplantation (AlloSCT) in paediatric recipients is associated with sustained engraftment and low risk of acute graft‐versus‐host disease (aGVHD), but limited by delayed immune reconstitution and increased risk of viral and fungal infection. The optimal dose of donor T cells to prevent graft failure and minimize risk of early opportunistic infection and post‐transplant lymphoproliferative disorder (PTLD), while avoiding severe aGVHD, remains unknown. We prospectively studied CD34‐selected 8–10/10 human leucocyte antigen (HLA)‐matched unrelated donor (MUD) peripheral blood stem cell transplantation (PBSCT) in a cohort of 19 paediatric AlloSCT recipients with malignant (n = 13) or non‐malignant (n = 6) diseases. T cells were added back to achieve total dose 1·0–2·5 × 105 CD3+/kg. GVHD pharmacoprophylaxis consisted only of tacrolimus. All patients engrafted neutrophils. Probabilities of grade II–IV aGVHD, limited chronic GVHD (cGVHD), and extensive cGVHD were 15·8%, 23·3%, and 0%, respectively. One patient developed PTLD. One‐year infection‐related mortality was 5·6%. T cell immune reconstitution was delayed. One‐year overall survival was 82·3%. Five patients with malignant disease ultimately died from progressive disease. CD34‐selected MUD PBSCT using a defined dose of T cell add‐back resulted in high rates of engraftment and low risk of grade II–IV aGVHD, early transplantation‐related mortality, and extensive cGVHD.


Cancer | 2017

Assessment of programmed death‐ligand 1 expression and tumor‐associated immune cells in pediatric cancer tissues

Robbie G. Majzner; Jason S. Simon; Joseph F. Grosso; Daniel Martinez; Bruce R. Pawel; Mariarita Santi; Melinda S. Merchant; Birgit Geoerger; Imene Hezam; Virginie Marty; Phillippe Vielh; Mads Daugaard; Poul H. Sorensen; Crystal L. Mackall; John M. Maris

Programmed death 1 (PD‐1) signaling in the tumor microenvironment dampens immune responses to cancer, and blocking this axis induces antitumor effects in several malignancies. Clinical studies of PD‐1 blockade are only now being initiated in pediatric patients, and little is known regarding programmed death‐ligand 1 (PD‐L1) expression in common childhood cancers. The authors characterized PD‐L1 expression and tumor‐associated immune cells (TAICs) (lymphocytes and macrophages) in common pediatric cancers.


Nature Medicine | 2018

Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M + diffuse midline gliomas

Christopher Mount; Robbie G. Majzner; Shree Sundaresh; Evan Arnold; Meena Kadapakkam; Samuel Haile; Louai Labanieh; Esther Hulleman; Pamelyn Woo; Skyler P. Rietberg; Hannes Vogel; Michelle Monje; Crystal L. Mackall

Diffuse intrinsic pontine glioma (DIPG) and other diffuse midline gliomas (DMGs) with mutated histone H3 K27M (H3-K27M)1–5 are aggressive and universally fatal pediatric brain cancers6. Chimeric antigen receptor (CAR)-expressing T cells have mediated impressive clinical activity in B cell malignancies7–10, and recent results suggest benefit in central nervous system malignancies11–13. Here, we report that patient-derived H3-K27M-mutant glioma cell cultures exhibit uniform, high expression of the disialoganglioside GD2. Anti-GD2 CAR T cells incorporating a 4-1BBz costimulatory domain14 demonstrated robust antigen-dependent cytokine generation and killing of DMG cells in vitro. In five independent patient-derived H3-K27M+ DMG orthotopic xenograft models, systemic administration of GD2-targeted CAR T cells cleared engrafted tumors except for a small number of residual GD2lo glioma cells. To date, GD2-targeted CAR T cells have been well tolerated in clinical trials15–17. Although GD2-targeted CAR T cell administration was tolerated in the majority of mice bearing orthotopic xenografts, peritumoral neuroinflammation during the acute phase of antitumor activity resulted in hydrocephalus that was lethal in a fraction of animals. Given the precarious neuroanatomical location of midline gliomas, careful monitoring and aggressive neurointensive care management will be required for human translation. With a cautious multidisciplinary clinical approach, GD2-targeted CAR T cell therapy for H3-K27M+ diffuse gliomas of pons, thalamus and spinal cord could prove transformative for these lethal childhood cancers.Lethal pediatric tumors bearing a particular histone H3 mutation upregulate the disialoganglioside GD2, thereby making these tumors susceptible to chimeric antigen receptor T cell–based immunotherapy.


Cancer Research | 2015

Abstract 249: Assessment of PD-L1 expression and tumor-associated lymphocytes in pediatric cancer tissues

Robbie G. Majzner; Jason S. Simon; Joseph F. Grosso; Daniel Martinez; Bruce R. Pawel; Mariarita Santi-Vincini; Melinda S. Merchant; Poul H. Sorensen; Crystal L. Mackall; John M. Maris

PD-1 signaling in the tumor microenvironment dampens immune responses to cancer and blocking this axis induces anti-tumor effects in several malignancies. Some studies have demonstrated increased efficacy of PD-1 blockade when tumor cells express PD-L1. Clinical studies of PD-1 blockade have not yet been conducted in pediatric patients and little is known regarding PD-L1 expression in common childhood cancers. We characterized PD-L1 expression and Tumor Associated Immune Cells (TAIC, lymphocytes and macrophages) in common pediatric cancers. Whole slide sections (n = 91) and tissue microarrays (n = 365) were evaluated by IHC for PD-L1 expression using the 28-8 anti-PD-L1 mAb in an automated Dako assay. PD-L1 expression was considered positive when at least 1% of tumor cells analyzed demonstrated plasma membrane staining. TAIC were also assessed for expression of PD-L1, and a subset of 60 tumors were assessed for CD3, CD4, CD8, CD45RO, PD-1, and FoxP3 expression on TAIC. Nine percent (N = 40) of evaluable tumors expressed PD-L1. Highest frequency histotypes comprised non-Hodgkin lymphoma (80%, 8/10), glioblastoma multiforme (30%, 6/20), and neuroblastoma (14%, 17/118). No PD-L1 staining was observed in Ewing sarcoma (0/20) or medulloblastoma (0/40). Despite a relatively low frequency of PD-L1+ pediatric cancers, the majority contained TAIC (73%, 334/456). Of these TAIC+ tumors, 73% of samples contained lymphocytes only, 25% contained lymphocytes and macrophages and 3% contained macrophages only. Twenty-one percent of TAIC+ samples demonstrated PD-L1 expression on TAIC, mostly comprising PD-L1 expression on macrophages (65%, 60/92), while lymphocytes expressed PD-L1 in only 7% (22/324). Taken together, PD-L1 was expressed in tumor and/or TAIC in 20% (90/456). Sixty samples were further analyzed to characterize TAIC, with 77% of the samples demonstrating infiltration of CD8+ T cells, most of which expressed CD45RO. Fox-P3 and PD-1 were expressed in 42% and 47% of the samples respectively. In summary, this provides the most comprehensive analysis of PD-L1 expression in pediatric associated cancer to date. Results show that a subset of pediatric cancers demonstrate tumor associated PD-L1 expression, while a much larger fraction demonstrate infiltration with tumor associated lymphocytes. Further preclinical and clinical investigation will define the predictive nature of PD-L1 expression in childhood cancers, but available data is not sufficient to exclude enrollment to clinical trials based on PD-L1 status. Citation Format: Robbie G. Majzner, Jason S. Simon, Joseph F. Grosso, Daniel Martinez, Bruce Pawel, Mariarita Santi-Vincini, Melinda S. Merchant, Poul Sorensen, Crystal L. Mackall, John M. Maris. Assessment of PD-L1 expression and tumor-associated lymphocytes in pediatric cancer tissues. [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 249. doi:10.1158/1538-7445.AM2015-249


Biology of Blood and Marrow Transplantation | 2017

Post-Transplantation Cyclophosphamide after Bone Marrow Transplantation Is Not Associated with an Increased Risk of Donor-Derived Malignancy

Robbie G. Majzner; Huzefa Mogri; Ravi Varadhan; Patrick Brown; Kenneth R. Cooke; Javier Bolaños-Meade; Lode J. Swinnen; Jennifer A. Kanakry; Leo Luznik; Richard J. Jones; Ephraim J. Fuchs; Rich Ambinder; Yvette L. Kasamon; Heather J. Symons

Post-transplantation cyclophosphamide (PTCy) can be used for graft-versus-host disease (GVHD) prophylaxis alone or in combination with other agents and is associated with excellent rates of engraftment and acute and chronic GVHD, as well as absence of post-transplantation lymphoproliferative disease. No study has previously evaluated the risk for developing donor-derived malignancy (DDM) in patients who receive PTCy. Giving chemotherapy in the immediate post-transplantation period carries with it a theoretic risk of disturbing the graft at a time of increased hematopoietic stress and causing or accelerating the development of malignancy. From 2000 to 2011, 789 patients underwent allogeneic transplantation and received PTCy at the Johns Hopkins Hospital. There were 4 cases of DDM identified among this large population, which is similar to or below the rate of DDM published in the literature. We found that the estimated cumulative incidence by competing risk analysis of DDM is 1.4% (SE, 1.02%). The use of PTCy does not appear to increase the risk of DDM.


Journal for ImmunoTherapy of Cancer | 2018

Durable regression of Medulloblastoma after regional and intravenous delivery of anti-HER2 chimeric antigen receptor T cells

Anandani Nellan; Christopher Rota; Robbie G. Majzner; Cynthia M. Lester-McCully; Andrea Griesinger; Jean M. Mulcahy Levy; Nicholas K. Foreman; Katherine E. Warren; Daniel W. Lee

BackgroundStandard-of-care therapies for treating pediatric medulloblastoma have long-term side effects, even in children who are cured. One emerging modality of cancer therapy that could be equally effective without such side effects would be chimeric antigen receptor (CAR) T cells. Knowing that human epidermal growth factor receptor 2 (HER2) is overexpressed in many medulloblastomas and has been used as a CAR T target before, we sought to evaluate the efficacy of more sophisticated anti-HER2 CAR T cells, as well as the feasibility and efficacy of different routes of delivering these cells, for the treatment of pediatric medulloblastoma.MethodsDaoy, D283 and D425 medulloblastoma cell lines were characterized by flow cytometry to evaluate HER2 expression. Anti-tumor efficacy of HER2-BBz-CAR T cells in vitro was performed using cytokine release and immune cytotoxicity assays compared to control CD19 CAR T cells. In vivo, Daoy and D283 tumor cells were orthotopically implanted in the posterior fossa of NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice and treated with regional or intravenous HER2-BBz-CAR T cells or control CD19 CAR T cells. Non-human primates (NHPs) bearing ventricular and lumbar reservoirs were treated with target autologous cells bearing extracellular HER2 followed by autologous HER2-CAR T cells intraventricularly. Cerebrospinal fluid and blood were collected serially to measure the persistence of delivered cells and cytokines.ResultsHER2-BBz-CAR T cells effectively clear medulloblastoma orthotopically implanted in the posterior fossa of NSG mice via both regional and intravenous delivery in xenograft models. Intravenous delivery requires a log higher dose compared to regional delivery. NHPs tolerated intraventricular delivery of autologous cells bearing extracellular HER2 followed by HER2-BBz-CAR T cells without experiencing any systemic toxicity.ConclusionsHER2-BBz-CAR T cells show excellent pre-clinical efficacy in vitro and in mouse medulloblastoma models, and their intraventricular delivery is feasible and safe in NHPs. A clinical trial of HER2-BBz-CAR T cells directly delivered into cerebrospinal fluid should be designed for patients with relapsed medulloblastoma.


Cancer Discovery | 2018

Tumor Antigen Escape from CAR T-cell Therapy

Robbie G. Majzner; Crystal L. Mackall

Emerging data from chimeric antigen receptor (CAR) T-cell trials in B-cell malignancies demonstrate that a common mechanism of resistance to this novel class of therapeutics is the emergence of tumors with loss or downregulation of the target antigen. Antigen loss or antigen-low escape is likely to emerge as an even greater barrier to success in solid tumors, which manifest greater heterogeneity in target antigen expression. Potential approaches to overcome this challenge include engineering CAR T cells to achieve multispecificity and to respond to lower levels of target antigen and more efficient induction of natural antitumor immune responses as a result of CAR-induced inflammation. In this article, we review the evidence to date for antigen escape and downregulation and discuss approaches currently under study to overcome these obstacles.Significance: Antigen escape and downregulation have emerged as major issues impacting the durability of CAR T-cell therapy. Here, we explore their incidence and ways to overcome these obstacles in order to improve clinical outcomes. Cancer Discov; 8(10); 1219-26. ©2018 AACR.


Nature Biomedical Engineering | 2018

Programming CAR-T cells to kill cancer

Louai Labanieh; Robbie G. Majzner; Crystal L. Mackall

T cells engineered to express chimeric antigen receptors (CARs) that are specific for tumour antigens have led to high complete response rates in patients with haematologic malignancies. Despite this early success, major challenges to the broad application of CAR-T cells as cancer therapies remain, including treatment-associated toxicities and cancer relapse with antigen-negative tumours. Targeting solid tumours with CAR-T cells poses additional obstacles because of the paucity of tumour-specific antigens and the immunosuppressive effects of the tumour microenvironment. To overcome these challenges, T cells can be programmed with genetic modules that increase their therapeutic potency and specificity. In this Review Article, we survey major advances in the engineering of next-generation CAR-T therapies for haematologic cancers and solid cancers, with particular emphasis on strategies for the control of CAR specificity and activity and on approaches for improving CAR-T-cell persistence and overcoming immunosuppression. We also lay out a roadmap for the development of off-the-shelf CAR-T cells.This Review Article provides an overview of chimeric antigen receptors (CARs) for T cells and discusses engineering strategies for the design of next-generation CAR-T therapies for haematologic and solid cancers.


Cancer Research | 2018

Abstract PR04: GD2-directed chimeric antigen receptor T cells mediate potent antitumor effect and cure in xenograft models of diffuse intrinsic pontine glioma

Robbie G. Majzner; Christopher Mount; Shree Sundaresh; Evan Arnold; Meena Kadapakkam; Louai Labanieh; Pamelyn Woo; Michelle Monje; Crystal L. Mackall

Introduction: Diffuse intrinsic pontine glioma (DIPG) is a universally fatal pediatric brainstem tumor with a median survival of less than one year. Despite advances in the understanding of the molecular origins of DIPG, improvement in clinical outcomes has yet to materialize. To date, there has been little target exploration for immunotherapy applications in DIPG. Methods: Patient-derived DIPG cell cultures were screened for expression of more than 350 surface antigens as potential immunotherapeutic targets. The disialoganglioside GD2 was found to have the highest expression across cell cultures and was verified by IHC on post-mortem samples. Chimeric antigen receptor (CAR) T-cell therapy against this target was explored both in vitro and in vivo. Results: We found high levels of the disialoganglioside GD2 expressed on cell cultures derived from post-mortem samples of DIPG. Quantification of the number of GD2 molecules per cell demonstrated higher GD2 expression on DIPG than any other tumors, including neuroblastoma, for which GD2 targeted immunotherapy is part of the standard of care. Most cases of DIPG are caused by a mutation in Histone 3.3 (H3K27M). GD2 is highly and uniformly expressed in patient-derived H3K27M DIPG cultures, whereas H3 wild-type pediatric high-grade gliomas, including those diagnosed as DIPG, do not express significant levels of GD2. The H3K27M mutation is associated with increased levels of enzymes in the ganglioside synthesis pathway, suggesting that expression of the target antigen is driven by H3K27M-induced transcriptional dysregulation. Anti-GD2 CAR T cells with a 4-1BB costimulatory domain demonstrate remarkable preclinical activity against H3K27M DIPG. GD2 CAR T cells specifically kill DIPG cells and produce cytokines IL-2 and IFN- upon coculture with tumor. Systemic administration of anti-GD2 CAR T cells achieves potent and durable cure compared to control T cells in multiple orthotopic xenograft models of DIPG. Using a CAR fluorescent protein fusion construct, we demonstrate significant T-cell trafficking to the brainstem where the antitumor effect is mediated. Universal response was observed across multiple cohorts, and treatment-associated toxicity was transient and tolerated during the period of peak antitumor activity. Conclusion: We have previously demonstrated that antigen density drives CAR efficacy. Extremely high expression of GD2 on DIPG makes this a particularly good disease for CAR T-cell therapy. If these results are predictive of human response, CAR T cells could have a transformative impact upon DIPG outcomes. A clinical trial of second generation anti-GD2 CAR T cells in relapsed and progressive DIPG is planned. Citation Format: Robbie G. Majzner, Christopher Mount, Shree Sundaresh, Evan Arnold, Meena Kadapakkam, Louai Labanieh, Pamelyn Woo, Michelle Monje, Crystal L. Mackall. GD2-directed chimeric antigen receptor T cells mediate potent antitumor effect and cure in xenograft models of diffuse intrinsic pontine glioma [abstract]. In: Proceedings of the AACR Special Conference: Pediatric Cancer Research: From Basic Science to the Clinic; 2017 Dec 3-6; Atlanta, Georgia. Philadelphia (PA): AACR; Cancer Res 2018;78(19 Suppl):Abstract nr PR04.

Collaboration


Dive into the Robbie G. Majzner'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