13 results on '"Meena Kadapakkam"'
Search Results
2. Data from Tuning the Antigen Density Requirement for CAR T-cell Activity
- Author
-
Crystal L. Mackall, Ronald D. Vale, Alexander R. Dunn, Peng Xu, Rachel C. Lynn, Johanna Theruvath, Volker Wiebking, Sang M. Nguyen, Sabine Heitzeneder, Rebecca M. Richards, Aidan M. Tousley, Evan W. Weber, Meena Kadapakkam, June H. Myklebust, Louai Labanieh, Vipul T. Vachharajani, Rui Dong, Elena Sotillo, Skyler P. Rietberg, and Robbie G. Majzner
- Abstract
Insufficient reactivity against cells with low antigen density has emerged as an important cause of chimeric antigen receptor (CAR) T-cell resistance. Little is known about factors that modulate the threshold for antigen recognition. We demonstrate that CD19 CAR activity is dependent upon antigen density and that the CAR construct in axicabtagene ciloleucel (CD19-CD28ζ) outperforms that in tisagenlecleucel (CD19-4-1BBζ) against antigen-low tumors. Enhancing signal strength by including additional immunoreceptor tyrosine-based activation motifs (ITAM) in the CAR enables recognition of low-antigen-density cells, whereas ITAM deletions blunt signal and increase the antigen density threshold. Furthermore, replacement of the CD8 hinge-transmembrane (H/T) region of a 4-1BBζ CAR with a CD28-H/T lowers the threshold for CAR reactivity despite identical signaling molecules. CARs incorporating a CD28-H/T demonstrate a more stable and efficient immunologic synapse. Precise design of CARs can tune the threshold for antigen recognition and endow 4-1BBζ-CARs with enhanced capacity to recognize antigen-low targets while retaining a superior capacity for persistence.Significance:Optimal CAR T-cell activity is dependent on antigen density, which is variable in many cancers, including lymphoma and solid tumors. CD28ζ-CARs outperform 4-1BBζ-CARs when antigen density is low. However, 4-1BBζ-CARs can be reengineered to enhance activity against low-antigen-density tumors while maintaining their unique capacity for persistence.This article is highlighted in the In This Issue feature, p. 627
- Published
- 2023
3. Tuning the Antigen Density Requirement for CAR T Cell Activity
- Author
-
Aidan Tousley, Sabine Heitzeneder, Johanna Theruvath, Robbie G. Majzner, Skyler P. Rietberg, Elena Sotillo, Volker Wiebking, Ronald D. Vale, Rui Dong, Rebecca Richards, Peng Xu, Evan W. Weber, Alexander R. Dunn, Rachel C. Lynn, Louai Labanieh, Vipul T. Vachharajani, Sang M. Nguyen, Meena Kadapakkam, Crystal L. Mackall, and June Helen Myklebust
- Subjects
0301 basic medicine ,Cell signaling ,Oncology and Carcinogenesis ,CD19 ,Article ,Synapse ,Vaccine Related ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Receptors ,medicine ,Animals ,Humans ,Tyrosine ,Cancer ,Receptors, Chimeric Antigen ,biology ,Chemistry ,Chimeric Antigen ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,Cell biology ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Immunization ,human activities ,CD8 ,Signal Transduction - Abstract
Insufficient reactivity against cells with low antigen density has emerged as an important cause of chimeric antigen receptor (CAR) T-cell resistance. Little is known about factors that modulate the threshold for antigen recognition. We demonstrate that CD19 CAR activity is dependent upon antigen density and that the CAR construct in axicabtagene ciloleucel (CD19-CD28ζ) outperforms that in tisagenlecleucel (CD19-4-1BBζ) against antigen-low tumors. Enhancing signal strength by including additional immunoreceptor tyrosine-based activation motifs (ITAM) in the CAR enables recognition of low-antigen-density cells, whereas ITAM deletions blunt signal and increase the antigen density threshold. Furthermore, replacement of the CD8 hinge-transmembrane (H/T) region of a 4-1BBζ CAR with a CD28-H/T lowers the threshold for CAR reactivity despite identical signaling molecules. CARs incorporating a CD28-H/T demonstrate a more stable and efficient immunologic synapse. Precise design of CARs can tune the threshold for antigen recognition and endow 4-1BBζ-CARs with enhanced capacity to recognize antigen-low targets while retaining a superior capacity for persistence. Significance: Optimal CAR T-cell activity is dependent on antigen density, which is variable in many cancers, including lymphoma and solid tumors. CD28ζ-CARs outperform 4-1BBζ-CARs when antigen density is low. However, 4-1BBζ-CARs can be reengineered to enhance activity against low-antigen-density tumors while maintaining their unique capacity for persistence. This article is highlighted in the In This Issue feature, p. 627
- Published
- 2020
4. A Novel Case of Carcinoid Tumor in a Pediatric Patient With Short Bowel Syndrome Secondary to Gastroschisis
- Author
-
Shweta S. Namjoshi, Raul Montiel-Esparza, Meena Kadapakkam, Alice C. Huang, Gregory Scott, Brock Martin, and Matias Bruzoni
- Subjects
Pediatric patient ,medicine.medical_specialty ,business.industry ,Gastroschisis ,Medicine ,business ,Short bowel syndrome ,medicine.disease ,Surgery - Published
- 2020
5. Repurposing Drugs for Acute Myeloid Leukemia: A Worthy Cause or a Futile Pursuit?
- Author
-
Anna V. Wojcicki, Adam Frymoyer, Kathleen M. Sakamoto, Hee-Don Chae, Meena Kadapakkam, and Norman J. Lacayo
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Review ,acute myeloid leukemia ,Malignancy ,lcsh:RC254-282 ,Unmet needs ,Food and drug administration ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Intensive care medicine ,Repurposing ,drug repurposing ,business.industry ,Myeloid leukemia ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,drug development ,targeted therapies ,Drug repositioning ,030104 developmental biology ,Oncology ,Drug development ,030220 oncology & carcinogenesis ,business ,Standard therapy ,mechanism of action - Abstract
Acute myeloid leukemia (AML) is a clinically and genetically heterogenous malignancy of myeloid progenitor cells that affects patients of all ages. Despite decades of research and improvement in overall outcomes, standard therapy remains ineffective for certain subtypes of AML. Current treatment is intensive and leads to a number of secondary effects with varying results by patient population. Due to the high cost of discovery and an unmet need for new targeted therapies that are well tolerated, alternative drug development strategies have become increasingly attractive. Repurposing existing drugs is one approach to identify new therapies with fewer financial and regulatory hurdles. In this review, we provide an overview of previously U.S. Food and Drug Administration (FDA) approved non-chemotherapy drugs under investigation for the treatment of AML.
- Published
- 2020
6. DIPG-36. ANTI-GD2 CHIMERIC ANTIGEN RECEPTOR T CELLS AS A POTENT IMMUNOTHERAPY REGIMEN IN XENOGRAFT MODELS OF HISTONE 3 K27M MUTANT DIFFUSE MIDLINE GLIOMA
- Author
-
Christopher Mount, Meena Kadapakkam, Michelle Monje, Samuel T. Haile, Louai Labanieh, Crystal L. Mackall, Pamelyn Woo, Evan Arnold, Skyler P. Rietberg, Shree Sundaresh, Hannes Vogel, and Robbie G. Majzner
- Subjects
Cancer Research ,biology ,Chemistry ,medicine.medical_treatment ,Mutant ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Histone H3 ,Abstracts ,Cytokine ,Histone ,Oncology ,Antigen ,Glioma ,Cancer research ,medicine ,biology.protein ,Neurology (clinical) - Abstract
Diffuse intrinsic pontine glioma (DIPG) and other histone H3 K27M (H3K27M) mutated diffuse midline gliomas (DMGs) are aggressive and universally fatal pediatric brain cancers. Chimeric antigen receptor (CAR)-expressing T-cells have mediated impressive clinical activity in B-cell malignancies, and recent results suggest benefit in CNS malignancies. Here, we report that patient-derived H3K27M-mutant glioma cell cultures exhibit uniform, high expression of the disialoganglioside GD2. Anti-GD2 CAR T-cells incorporating a 4-1BBz costimulatory domain demonstrated robust antigen-dependent cytokine generation and DMG cell killing in vitro. In five independent patient-derived H3K27M+ DMG orthotopic xenograft models, systemic administration of GD2-CAR T-cells cleared engrafted tumors, save a small number of residual GD2-low glioma cells. Using a fluorescently-labeled GD2-CAR, we demonstrate tumoricidal GD2-CAR T cells infiltrating the brain parenchyma and sparing of local neurons during tumor clearing. While GD2-CAR T-cell administration was tolerated in the majority of animals, peri-tumoral neuroinflammation during the acute phase of antitumor activity resulted in hydrocephalus that was lethal in a fraction of animals. Similar swelling in thalamic xenograft models is lethal in a significant fraction of animals, presumably due to third ventricular compression and lethal transtentorial herniation. 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-CAR T-cell therapy for H3K27M+ diffuse gliomas of the pons, thalamus and spinal cord could prove transformative for these lethal childhood cancers.
- Published
- 2018
7. Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M+ diffuse midline gliomas letter
- Author
-
Esther Hulleman, Pamelyn Woo, Hannes Vogel, Robbie G. Majzner, Louai Labanieh, Samuel T. Haile, Michelle Monje, Christopher Mount, Evan Arnold, Meena Kadapakkam, Crystal L. Mackall, Skyler P. Rietberg, Shree Sundaresh, Pediatric surgery, and CCA - Cancer biology and immunology
- Subjects
0301 basic medicine ,business.industry ,medicine.medical_treatment ,Central nervous system ,General Medicine ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Chimeric antigen receptor ,In vitro ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Glioma ,medicine ,Systemic administration ,Cancer research ,business ,Neuroinflammation ,B cell - Abstract
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 cancers 6 . Chimeric antigen receptor (CAR)-expressing T cells have mediated impressive clinical activity in B cell malignancies 7-10, and recent results suggest benefit in central nervous system malignancies 11-13 . 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 domain 14 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 trials 15-17 . 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.
- Published
- 2018
8. Phase I Trial Using CD19/CD22 Bispecific CAR T Cells in Pediatric and Adult Acute Lymphoblastic Leukemia (ALL)
- Author
-
Anne Cunniffe Marcy, Terry J. Fry, Bita Sahaf, Kara L. Davis, Crystal L. Mackall, Juliana Craig, Michelle Fujimoto, Lori Muffly, Haiying Qin, Katherine A. Kong, Nasheed Hossain, Jay Y. Spiegel, Jenny Sumin Yoon, David B. Miklos, Robbie G. Majzner, Liora M. Schultz, Emily Egeler, Neehar Bhatia, Sneha Ramakrishna, Meena Kadapakkam, Christina Baggott, Courtney Erickson, Sharon Mavroukakis, Everett Meyer, Matthew J. Frank, Shabnum Patel, and Steven A. Feldman
- Subjects
Oncology ,medicine.medical_specialty ,Cyclophosphamide ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Fludarabine ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,Acute lymphocytic leukemia ,medicine ,Adult Acute Lymphoblastic Leukemia ,Blinatumomab ,Bone marrow ,business ,medicine.drug - Abstract
Introduction: Chimeric antigen receptor (CAR) T cells targeting either CD19 or CD22 have yielded striking complete remission (CR) rates of 70%-90% in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (ALL), but CD19 negative and CD22 low relapse limits the curative potential of these single-antigen CAR T cell approaches. We hypothesized that a bivalent CAR-T construct that can target CD19 and/or CD22 would prevent antigen negative/low relapse. Here we present the combined single institution experience to date of pediatric and adult patients with R/R ALL treated with this novel bispecific CAR. Methods: We conducted parallel Phase I clinical trials of CD19/CD22 bispecific CAR T cells in pediatric and adult patients with relapsed/refractory ALL. We utilized lentiviral transduction of a bivalent CAR construct incorporating the fmc63 CD19 and m971 CD22 single chain variable fragments (scFvs) and a 41BB costimulatory endodomain. After lymphodepletion with fludarabine and cyclophosphamide, patients were infused with fresh or cryopreserved CAR T cells manufactured using a 7-11 day process. Two dose levels were tested during dose escalation: Dose level 1 was 1x106 CAR T cells/kg and dose level 2 was 3x106 cells/kg. Primary objectives assessed the ability to successfully manufacture CAR19/22 CAR T cells and safety while response at Day 28 post-infusion was a secondary objective. Blood, bone marrow and cerebrospinal fluid samples were obtained at protocol defined intervals for correlative biology studies. Results: Nineteen patients have been enrolled (10 pediatric; 9 adult) with a median age of 23 years (range, 2-68) and median of 4 (range, 2-11) prior lines of leukemia-directed therapy. Ten patients received prior HCT, 9 were treated with prior Blinatumomab, 3 with prior CD19 directed CAR T cells and 4 with prior Inotuzumab. Fourteen patients (8 pediatric, 6 adult) have been infused to date with CD19/CD22 bispecific CAR T cells; 7 were treated at dose level 1 (DL1) and 7 at dose level 2 (DL2). Successful manufacturing of cells at target dose levels was achieved in all patients. Twelve patients have reached day 28 and are included in the safety and response analysis presented here. Nine of 12 (75%) experienced cytokine release syndrome (CRS) and 2/12 (17%) developed immune-effector cell neurotoxicity syndrome (ICANS). The CRS and ICANS were all grade 1 or 2 across both dose levels and across pediatric and adult patients except for one adult with high disease burden who experienced grade 4 CRS and grade 4 ICANS, both of which were reversible. No differences in toxicities were seen across the patient age spectrum and there were no cases of treatment-related mortality within 28 days following CAR T infusion. Eleven of 12 (92%) patients achieved a CR, 10 of whom achieved CR at day 28 and one with a PR of extramedullary disease at day 28 which improved to CR by day 180 without further leukemia-directed intervention. One patient had primary progressive disease prior to day 28. Peak CAR expansion as detected by peripheral blood flow cytometry reached a median level of 11.13% (DL1) and 29.1% (DL2) CAR T of CD3+ cells with a range of 0.7-22.54% and 3.8-86.96%, respectively. To date, 3 patients (1 pediatric and 2 adult patients) have relapsed, all with retention of CD19. Post-remission practice differed across pediatric and adult patients; Six pediatric patients reaching day 28 underwent consolidative hematopoietic cell transplantation (HCT) whereas no adult patients received subsequent HCT. One patient died from complications post HCT while in remission. Therefore, the overall survival for all infused patients was 92% with a median follow-up of 9.5 months from time of infusion (range, 1-20). Conclusion: The combined pediatric and adult phase I trials of bispecific CD19/CD22 targeting CAR T cells in relapsed/refractory ALL demonstrates safety and tolerability at two dose levels. Expanded accrual at dose level 2 is ongoing and clinical outcomes will be updated. This work additionally demonstrates feasibility of delivering unified B-ALL CAR T cell therapy across age boundaries. Multi-parametric CyTOF studies permitting CAR T cell phenotyping in conjunction with single cell TCR tracking, proteomics, epigenomics and cytokine profiling are ongoing and will be used to further characterize persisting CAR T cells and define inter-product and inter-patient variability. Disclosures Muffly: Pfizer: Consultancy; KITE: Consultancy; Adaptive: Research Funding. Majzner:Xyphos Inc.: Consultancy; Lyell Immunopharma: Consultancy. Feldman:Octane Biotech, Inc.: Employment; Personalized Medicine Initiative Science: Membership on an entity's Board of Directors or advisory committees. Miklos:Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; Kite-Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Juno: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Becton Dickinson: Research Funding; Miltenyi Biotech: Membership on an entity's Board of Directors or advisory committees; Precision Bioscience: Membership on an entity's Board of Directors or advisory committees; AlloGene: Membership on an entity's Board of Directors or advisory committees. Mackall:Obsidian: Research Funding; Lyell: Consultancy, Equity Ownership, Other: Founder, Research Funding; Nektar: Other: Scientific Advisory Board; PACT: Other: Scientific Advisory Board; Bryologyx: Other: Scientific Advisory Board; Vor: Other: Scientific Advisory Board; Roche: Other: Scientific Advisory Board; Adaptimmune LLC: Other: Scientific Advisory Board; Glaxo-Smith-Kline: Other: Scientific Advisory Board; Allogene: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Apricity Health: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Unum Therapeutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2019
9. Constitutional tandem duplication of 9q34 that truncates EHMT1 in a child with ganglioglioma
- Author
-
Hélène Legay, Svetlana A. Yatsenko, Meena Kadapakkam, James R. Lupski, Hannah C. Cheung, Sharon E. Plon, and Jack Su
- Subjects
Genetics ,Breakpoint ,Intron ,Histone-Lysine N-Methyltransferase ,Hematology ,Biology ,TNF Receptor-Associated Factor 2 ,medicine.disease ,Article ,Ganglioglioma ,Oncology ,Fusion transcript ,Child, Preschool ,Chromosome Duplication ,Pediatrics, Perinatology and Child Health ,Speech delay ,Gene duplication ,medicine ,Humans ,Female ,Tandem exon duplication ,medicine.symptom ,Chromosomes, Human, Pair 9 ,Anaplastic astrocytoma - Abstract
Point mutations of EHMT1 or deletions and duplications of chromosome 9q34.3 are found in patients with variable neurologic and developmental disorders. Here, we present a child with congenital cataract, developmental and speech delay who developed a metastatic ganglioglioma with progression to anaplastic astrocytoma. Molecular analysis identified a novel constitutional tandem duplication in 9q34.3 with breakpoints in intron 1 of TRAF2 and intron 16 of EHMT1 generating a fusion transcript predicted to encode a truncated form of EHMT1. The ganglioglioma showed complex chromosomal aberrations with further duplication of the dup9q34. Thus, this unique tandem 9q34.3 duplication may impact brain tumor formation.
- Published
- 2011
10. Abstract PR04: GD2-directed chimeric antigen receptor T cells mediate potent antitumor effect and cure in xenograft models of diffuse intrinsic pontine glioma
- Author
-
Christopher Mount, Crystal L. Mackall, Shree Sundaresh, Pamelyn Woo, Evan Arnold, Michelle Monje, Robbie G. Majzner, Louai Labanieh, and Meena Kadapakkam
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,Cell ,Cancer ,Immunotherapy ,medicine.disease ,Pediatric cancer ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,Antigen ,Cell culture ,Neuroblastoma ,Cancer research ,Medicine ,business - Abstract
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.
- Published
- 2018
11. Visceral primary non-rhabdomyosarcoma soft tissue sarcoma (NRSTS) in patients < 30 years of age: Findings of Children’s Oncology Group (COG) study ARST0332
- Author
-
Mary Beth McCarville, Jingying Weng, Donald A. Barkauskas, Lynn Million, Douglas S. Hawkins, Sheri L. Spunt, Susan M. Hiniker, and Meena Kadapakkam
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Soft tissue sarcoma ,medicine.disease ,Optimal management ,Cog ,Internal medicine ,medicine ,In patient ,business ,Rhabdomyosarcoma - Abstract
10545Background: Little is known about the clinical features, optimal management, and prognosis of young patients with primary visceral NRSTS. Methods: We analyzed clinical features, treatment, and...
- Published
- 2018
12. PEGylation strategies for active targeting of PLA/PLGA nanoparticles
- Author
-
James D. Byrne, Nicole Sunaryo, Tania Betancourt, Lisa Brannon-Peppas, Shefali Patel, Spencer W. Crowder, Shelly L. Casciato, and Meena Kadapakkam
- Subjects
Immunoconjugates ,Materials science ,Polymers ,Surface Properties ,Polyesters ,Biomedical Engineering ,Poloxamer ,macromolecular substances ,Polyethylene Glycols ,Biomaterials ,Mice ,chemistry.chemical_compound ,Drug Delivery Systems ,Polylactic Acid-Polyglycolic Acid Copolymer ,PEG ratio ,Polymer chemistry ,medicine ,Animals ,Lactic Acid ,Carbodiimide ,Lactide ,technology, industry, and agriculture ,Metals and Alloys ,PLGA ,chemistry ,Chemical engineering ,Poloxamer 407 ,Ceramics and Composites ,PEGylation ,Nanoparticles ,Ethylene glycol ,Polyglycolic Acid ,medicine.drug - Abstract
This work evaluates various techniques for the incorporation of poly(ethylene glycol) (PEG) onto biodegradable nanoparticles (NPs) of poly(lactic-co-glycolic acid) (PLGA) or poly(lactic acid) (PLA) with the purpose of providing a functional site for surface conjugation of targeting agents and for improving surface properties. The techniques compared were based on NP preparation with blends of PLGA and poloxamer or with block copolymers of PLGA/PLA with PEG. Blending of PLGA with poloxamer 407 resulted in the incorporation of the latter to up to a 43 wt % content. Direct conjugation of heterofunctional NH2-PEG-COOH to the surface of premade NPs was not highly effective. Preparation of copolymers of PLGA with PEG was determined to be more effective and versatile by polymerization of lactide and glycolide dimers onto the hydroxyl group of heterofunctional OH-PEG-COOH than by conjugation of the premade polymers with carbodiimide chemistry. NPs prepared with these copolymers confirmed the surface localization of PEG and proved to be useful for conjugation of mouse immumoglobulin as a model targeting agent.
- Published
- 2009
13. PDTM-39. GD2-DIRECTED CHIMERIC ANTIGEN RECEPTOR T CELLS AS A POTENT IMMUNOTHERAPY REGIMEN IN XENOGRAFT MODELS OF DIFFUSE INTRINSIC PONTINE GLIOMA
- Author
-
Meena Kadapakkam, Michelle Monje-Deisseroth, Christopher Mount, Shree Sundaresh, Robbie G. Majzner, Evan Arnold, and Crystal L. Mackall
- Subjects
0301 basic medicine ,Cancer Research ,Mutation ,Chemistry ,medicine.medical_treatment ,Treatment outcome ,Immunotherapy ,medicine.disease_cause ,Chimeric antigen receptor ,Pons ,Abstracts ,03 medical and health sciences ,Regimen ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Cancer research ,Neurology (clinical) - Abstract
Diffuse intrinsic pontine glioma (DIPG) is a universally fatal pediatric brainstem tumor with a median survival of approximately 10 months. Despite advances in understanding the molecular origins of DIPG, translations to improvement in clinical outcomes have yet to materialize. To date, there has been little target exploration for immunotherapy applications in DIPG. Here we report substantial preclinical efficacy of chimeric antigen receptor (CAR)-bearing T cells targeting the disialoganglioside GD2 as an immunotherapy regimen in patient-derived orthotopic xenograft models of Histone 3 K27M (H3K27M) mutated DIPG. GD2 is highly and uniformly expressed in patient-derived H3K27M DIPG cultures, and in vitro assays demonstrate substantial target-dependent cytokine generation and target cell killing. H3 WT pediatric high-grade gliomas (HGGs), including one case diagnosed as DIPG, do not express significant levels of GD2, and the H3K27M mutation is associated with increased synthesis of ganglioside pathway synthesis enzymes, suggesting expression of the GD2 antigen is driven by H3K27M-induced transcriptional dysregulation. Single-dose systemic administration of GD2-directed CAR T cells in multiple orthotopic xenograft models of DIPG achieves potent and lasting antitumor efficacy within a 4 week period of administration compared to a CD19-directed CAR control cohort. Universal response was observed across multiple cohorts, and treatment-associated toxicity was transient and tolerated during the period of peak anti-tumor activity. If these results are predictive of human response, GD2-directed CAR T cell therapy could have a transformative impact upon DIPG outcomes.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.