51
|
Liu R, Mitchell DA. Survivin as an immunotherapeutic target for adult and pediatric malignant brain tumors. Cancer Immunol Immunother 2010; 59:183-93. [PMID: 19756596 PMCID: PMC11030045 DOI: 10.1007/s00262-009-0757-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 08/18/2009] [Indexed: 12/22/2022]
Affiliation(s)
- Rebecca Liu
- The Preston Robert Tisch Brain Tumor Center at Duke, Durham, NC, USA.
| | | |
Collapse
|
52
|
Abstract
Over the past decade, dendritic cell-based immunotherapy for central nervous system tumors has progressed from preclinical rodent models and safety assessments to phase I/II clinical trials in over 200 patients, which have produced measurable immunologic responses and some prolonged survival rates. Many questions regarding the methods and molecular mechanisms behind this new treatment option, however, remain unanswered. Results from currently ongoing and future studies will help to elucidate which dendritic cell preparations, treatment protocols, and adjuvant therapeutic regimens will optimize the efficacy of dendritic cell vaccination. As clinical studies continue to report results on dendritic cell-mediated immunotherapy, it will be critical to continue refining treatment methods and developing new ways to augment this promising form of glioma treatment.
Collapse
Affiliation(s)
- Won Kim
- UCLA Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California, 90095
| | - Linda M. Liau
- UCLA Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California, 90095
| |
Collapse
|
53
|
Kateb B, Yamamoto V, Alizadeh D, Zhang L, Manohara HM, Bronikowski MJ, Badie B. Multi-walled carbon nanotube (MWCNT) synthesis, preparation, labeling, and functionalization. Methods Mol Biol 2010; 651:307-317. [PMID: 20686974 DOI: 10.1007/978-1-60761-786-0_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Nanomedicine is a growing field with a great potential for introducing new generation of targeted and personalized drug. Amongst new generation of nano-vectors are carbon nanotubes (CNTs), which can be produced as single or multi-walled. Multi-walled carbon nanotubes (MWCNTs) can be fabricated as biocompatible nanostructures (cylindrical bulky tubes). These structures are currently under investigation for their application in nanomedicine as viable and safe nanovectors for gene and drug delivery. In this chapter, we will provide you with the necessary information to understand the synthesis of MWCNTs, functionalization, PKH26 labeling, RNAi, and DNA loading for in vitro experimentation and in vivo implantation of labeled MWCNT in mice as well as materials used in this experimentation. We used this technique to manipulate microglia as part of a novel application for the brain cancer immunotherapy. Our published data show this is a promising technique for labeling, and gene and drug delivery into microglia.
Collapse
Affiliation(s)
- Babak Kateb
- Brain Mapping Foundation and International Brain Mapping & Intraoperative Surgical Planning Society (IBMISPS), West Hollywood, CA, USA
| | | | | | | | | | | | | |
Collapse
|
54
|
Abstract
This article provides a broad overview of the data, including laboratory and clinical studies, currently available on the combination of immunotherapy and chemotherapy for treating cancer. The various forms of immunotherapy combined with chemotherapy include monoclonal antibodies, adoptive lymphocyte transfer, or active specific immunotherapy, such as tumor proteins, irradiated tumor cells, tumor cell lysates, dendritic cells pulsed with peptides or lysates, or tumor antigens expressed in plasmids or viral vectors. This discussion is not limited to malignant brain tumors, because many of the studies have been conducted on various cancer types, thereby providing a comprehensive perspective that may encourage further studies that combine chemotherapy and immunotherapy for treating brain tumors.
Collapse
|
55
|
Abstract
Several immunostimulant approaches have been studied in the treatment of gliomas. The advent of recombinant DNA technology led to a nonspecific immunostimulation via systemic administration of cytokines. Recently, in attempts to more closely mimic their natural activity, cytokines have been delivered by implanting genetically transduced cells or by using in vivo gene transfer techniques. The latest efforts have focused on immunostimulatory agents that act directly on antigen-presenting cells and effector cells of the immune system via pattern recognition receptors. Combining these strategies with more than one mode of immunotherapy may provide better clinical results.
Collapse
Affiliation(s)
- Nicholas Butowski
- Department of Neurological Surgery, University of California San Francisco, 400 Parnassus Avenue, A808, San Francisco, CA 94143, USA.
| |
Collapse
|
56
|
Agarwalla PK, Barnard ZR, Curry WT. Virally mediated immunotherapy for brain tumors. Neurosurg Clin N Am 2009; 21:167-79. [PMID: 19944975 DOI: 10.1016/j.nec.2009.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Brain tumors are a leading cause of mortality and morbidity in the United States. Malignant brain tumors occur in approximately 80,000 adults. Furthermore, the average 5-year survival rate for malignant brain tumors across all ages and races is approximately 30% and has remained relatively static over the past few decades, showing the need for continued research and progress in brain tumor therapy. Improved techniques in molecular biology have expanded understanding of tumor genetics and permitted viral engineering and the anticancer therapeutic use of viruses as directly cytotoxic agents and as gene vectors. Preclinical models have shown promising antitumor effects, and generation of clinical grade vectors is feasible. In parallel to these developments, better understanding of antitumor immunity has been accompanied by progress in cancer immunotherapy, the goal of which is to stimulate host rejection of a growing tumor. This article reviews the intersection between the use of viral therapy and immunotherapy in the treatment of malignant gliomas. Each approach shows great promise on its own and in combined or integrated forms.
Collapse
Affiliation(s)
- Pankaj K Agarwalla
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
57
|
Abstract
Glioblastoma multiforme is the most common primary central nervous system tumor. The prognosis for these malignant brain tumors is poor, with a median survival of 14 months and a 5-year survival rate below 2%. Development of novel treatments is essential to improving survival and quality of life for these patients. Endogenous heat shock proteins have been implicated in mediation of both adaptive and innate immunity, and there is a rising interest in the use of this safe and multifaceted heat shock protein vaccine therapy as a promising treatment for human cancers, including glioblastoma multiforme.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurological Surgery, University of California at San Francisco, 505 Parnassus Avenue, Room M779, Campus 0112, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
58
|
Mitra S, Li G, Harsh GR. Passive antibody-mediated immunotherapy for the treatment of malignant gliomas. Neurosurg Clin N Am 2009; 21:67-76. [PMID: 19944967 DOI: 10.1016/j.nec.2009.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Despite advances in understanding the molecular mechanisms of brain cancer, the outcome of patients with malignant gliomas treated according to the current standard of care remains poor. Novel therapies are needed, and immunotherapy has emerged with great promise. The diffuse infiltration of malignant gliomas is a major challenge to effective treatment; immunotherapy has the advantage of accessing the entire brain with specificity for tumor cells. Therapeutic immune approaches include cytokine therapy, passive immunotherapy, and active immunotherapy. Cytokine therapy involves the administration of immunomodulatory cytokines to activate the immune system. Active immunotherapy is the generation or augmentation of an immune response, typically by vaccination against tumor antigens. Passive immunotherapy connotes either adoptive therapy, in which tumor-specific immune cells are expanded ex vivo and reintroduced into the patient, or passive antibody-mediated therapy. In this article, the authors discuss the preclinical and clinical studies that have used passive antibody-mediated immunotherapy, otherwise known as serotherapy, for the treatment of malignant gliomas.
Collapse
Affiliation(s)
- Siddhartha Mitra
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, Edwards Building Room 200, Stanford, CA 94305, USA
| | | | | |
Collapse
|
59
|
The role of tregs in glioma-mediated immunosuppression: potential target for intervention. Neurosurg Clin N Am 2009; 21:125-37. [PMID: 19944972 DOI: 10.1016/j.nec.2009.08.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of regulatory T cells (Tregs) in mediating immune suppression of anti-tumor immune responses is increasingly appreciated in patients with malignancies-especially within the malignant glioma patient population. This article discuss the role and prognostic significance of Tregs within glioma patients and delineates potential approaches for their inhibition that can be used alone or in combination with other immune therapeutics in clinical trials and in the clinical settings of recurrent or residual disease.
Collapse
|
60
|
Kanaly CW, Ding D, Heimberger AB, Sampson JH. Clinical applications of a peptide-based vaccine for glioblastoma. Neurosurg Clin N Am 2009; 21:95-109. [PMID: 19944970 DOI: 10.1016/j.nec.2009.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glioblastoma multiforme is a malignant, relentless brain cancer with no known cure, and standard therapies leave significant room for the development of better, more effective treatments. Immunotherapy is a promising approach to the treatment of solid tumors that directs the patient's own immune system to destroy tumor cells. The most successful immunologically based cancer therapy to date involves the passive administration of monoclonal antibodies, but significant antitumor responses have also been generated with active vaccination strategies and cell-transfer therapies. This article summarizes the important components of the immune system, discusses the specific difficulty of immunologic privilege in the central nervous system, and reviews treatment approaches that are being attempted, with an emphasis on active immunotherapy using peptide vaccines.
Collapse
Affiliation(s)
- Charles W Kanaly
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Box 3050, 220 Sands Building, Research Drive, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
61
|
Zhang L, Alizadeh D, Van Handel M, Kortylewski M, Yu H, Badie B. Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice. Glia 2009; 57:1458-67. [PMID: 19306372 DOI: 10.1002/glia.20863] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
As the main effector-cell population of the central nervous system, microglia (MG) are considered to play an important immunoregulatory function in a number of pathological conditions such as inflammation, trauma, degenerative disease, and brain tumors. Recent studies, however, have suggested that the anti-neoplastic function of MG may be suppressed in malignant brain tumors. Considering the proposed suppressive role of signal transducers and activators of transcription 3 (Stat3) in antitumor immunity, we evaluated the role of Stat3 inhibition on MG and macrophage (MP) activation and tumor growth in a murine glioma model. N9 MG cells were exposed to GL261 glioma conditioned medium (GL261-CM) and evaluated for Stat3 activity and cytokine expression. Furthermore, the role of Stat3 inhibition on MG and MP activation was studied both in vitro and in vivo. Finally, the effect of Stat3 inhibition on tumor growth was assessed in intracranial GL261 gliomas. GL261-CM increased Stat3 activity in N9 cells in vitro and resulted in overexpression of IL-10 and IL-6, and downregulation of IL1-beta, a pro-inflammatory cytokine. Inhibition of Stat3 by CPA-7 or siRNA reversed glioma-induced cytokine expression profile in N9 cells. Furthermore, inactivation of Stat3 in intracranial GL261 tumors by siRNA resulted in MG/MP activation and tumor growth inhibition. Glioma-induced MG and MP suppression may be mediated thorough Stat3. Inhibition of Stat3 function in tumor MG/MP may result in their activation and can potentially be used as an adjunct immunotherapy approach for gliomas.
Collapse
Affiliation(s)
- Leying Zhang
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, Duarte, California, USA
| | | | | | | | | | | |
Collapse
|
62
|
Choi BD, Archer GE, Mitchell DA, Heimberger AB, McLendon RE, Bigner DD, Sampson JH. EGFRvIII-targeted vaccination therapy of malignant glioma. Brain Pathol 2009; 19:713-23. [PMID: 19744042 PMCID: PMC2846812 DOI: 10.1111/j.1750-3639.2009.00318.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 06/30/2009] [Indexed: 12/25/2022] Open
Abstract
Given the highly infiltrative growth pattern of malignant glioma and the lack of specificity associated with currently available treatment regimens, alternative strategies designed to eradicate cancer cells while limiting collateral toxicity in normal tissues remain a high priority. To this end, the development of specific immunotherapies against targeted neoplastic cells represents a promising approach. The epidermal growth factor receptor class III variant (EGFRvIII), a constitutively activated mutant of the wild-type tyrosine kinase, is present in a substantial proportion of malignant gliomas and other human cancers, yet completely absent from normal tissues. This receptor variant consists of an in-frame deletion, the translation of which produces an extracellular junction with a novel glycine residue, flanked by amino acid sequences that are not typically adjacent in the normal protein. In this review, both preclinical and early clinical development of a peptide vaccine directed against this portion of the EGFRvIII antigenic domain are recapitulated. Following vaccination, our group has demonstrated potent, redirected cellular and humoral immunity against cancer cells expressing the mutant receptor without significant toxicity. Additionally, the corresponding therapeutic outcomes observed in these studies lend credence to the potential role of peptide-based vaccination strategies among emerging antitumor immunotherapies in patients with malignant glioma.
Collapse
Affiliation(s)
- Bryan D Choi
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | |
Collapse
|
63
|
Ghulam Muhammad AKM, Candolfi M, King GD, Yagiz K, Foulad D, Mineharu Y, Kroeger KM, Treuer KA, Nichols WS, Sanderson NS, Yang J, Khayznikov M, Van Rooijen N, Lowenstein PR, Castro MG. Antiglioma immunological memory in response to conditional cytotoxic/immune-stimulatory gene therapy: humoral and cellular immunity lead to tumor regression. Clin Cancer Res 2009; 15:6113-27. [PMID: 19789315 DOI: 10.1158/1078-0432.ccr-09-1087] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastoma multiforme is a deadly primary brain cancer. Because the tumor kills due to recurrences, we tested the hypothesis that a new treatment would lead to immunological memory in a rat model of recurrent glioblastoma multiforme. EXPERIMENTAL DESIGN We developed a combined treatment using an adenovirus (Ad) expressing fms-like tyrosine kinase-3 ligand (Flt3L), which induces the infiltration of immune cells into the tumor microenvironment, and an Ad expressing herpes simplex virus-1-thymidine kinase (TK), which kills proliferating tumor cells in the presence of ganciclovir. RESULTS This treatment induced immunological memory that led to rejection of a second glioblastoma multiforme implanted in the contralateral hemisphere and of an extracranial glioblastoma multiforme implanted intradermally. Rechallenged long-term survivors exhibited anti-glioblastoma multiforme-specific T cells and displayed specific delayed-type hypersensitivity. Using depleting antibodies, we showed that rejection of the second tumor was dependent on CD8(+) T cells. Circulating anti-glioma antibodies were observed when glioblastoma multiforme cells were implanted intradermally in naïve rats or in long-term survivors. However, rats bearing intracranial glioblastoma multiforme only exhibited circulating antitumoral antibodies upon treatment with Ad-Flt3L + Ad-TK. This combined treatment induced tumor regression and release of the chromatin-binding protein high mobility group box 1 in two further intracranial glioblastoma multiforme models, that is, Fisher rats bearing intracranial 9L and F98 glioblastoma multiforme cells. CONCLUSIONS Treatment with Ad-Flt3L + Ad-TK triggered systemic anti-glioblastoma multiforme cellular and humoral immune responses, and anti-glioblastoma multiforme immunological memory. Release of the chromatin-binding protein high mobility group box 1 could be used as a noninvasive biomarker of therapeutic efficacy for glioblastoma multiforme. The robust treatment efficacy lends further support to its implementation in a phase I clinical trial.
Collapse
Affiliation(s)
- A K M Ghulam Muhammad
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Kennedy BC, Maier LM, D'Amico R, Mandigo CE, Fontana EJ, Waziri A, Assanah MC, Canoll P, Anderson RCE, Anderson DE, Bruce JN. Dynamics of central and peripheral immunomodulation in a murine glioma model. BMC Immunol 2009; 10:11. [PMID: 19226468 PMCID: PMC2654428 DOI: 10.1186/1471-2172-10-11] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Accepted: 02/18/2009] [Indexed: 11/26/2022] Open
Abstract
Background Immunosuppression by gliomas contributes to tumor progression and treatment resistance. It is not known when immunosuppression occurs during tumor development but it likely involves cross-talk among tumor cells, tumor-associated macrophages and microglia (TAMs), and peripheral as well as tumor-infiltrating lymphocytes (TILs). Results We have performed a kinetic study of this immunomodulation, assessing the dynamics of immune infiltration and function, within the central nervous system (CNS) and peripherally. PDGF-driven murine glioma cells were injected into the white matter of 13 mice. Four mice were sacrificed 13 days post-injection (dpi), four mice at 26 dpi, and five mice at 40 dpi. Using multiparameter flow cytometry, splenic T cells were assessed for FoxP3 expression to identify regulatory T cells (Tregs) and production of IFN-γ and IL-10 after stimulation with PMA/ionomycin; within the CNS, CD4+ TILs were quantified, and TAMs were quantified and assessed for TNF-α and IL-10 production after stimulation with LPS. Peripheral changes associated with tumor development were noted prior to effects within the CNS. The percentage of FoxP3+ regulatory T cells (Tregs) increased by day 26, with elevated frequencies throughout the duration of the study. This early increase in Tregs was paralleled by an increase in IL-10 production from Tregs. At the final time points examined (tumor morbidity or 40 dpi), there was an increase in the frequency of TAMs with decreased capacity to secrete TNF-α. An increase in TIL frequency was also observed at these final time points. Conclusion These data provide insight into the kinetics of the immunosuppressive state associated with tumor growth in a murine model of human gliomas. Functional impairment of TAMs occurs relatively late in the course of GBM tumor growth, potentially providing a window of opportunity for therapeutic strategies directed towards preventing their functional impairment.
Collapse
Affiliation(s)
- Benjamin C Kennedy
- Department of Neurosurgery, Gabriele Bartoli Brain Tumor Research Laboratory Columbia University, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Curtin JF, Liu N, Candolfi M, Xiong W, Assi H, Yagiz K, Edwards MR, Michelsen KS, Kroeger KM, Liu C, Muhammad AKMG, Clark MC, Arditi M, Comin-Anduix B, Ribas A, Lowenstein PR, Castro MG. HMGB1 mediates endogenous TLR2 activation and brain tumor regression. PLoS Med 2009; 6:e10. [PMID: 19143470 PMCID: PMC2621261 DOI: 10.1371/journal.pmed.1000010] [Citation(s) in RCA: 277] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Accepted: 11/19/2008] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor that carries a 5-y survival rate of 5%. Attempts at eliciting a clinically relevant anti-GBM immune response in brain tumor patients have met with limited success, which is due to brain immune privilege, tumor immune evasion, and a paucity of dendritic cells (DCs) within the central nervous system. Herein we uncovered a novel pathway for the activation of an effective anti-GBM immune response mediated by high-mobility-group box 1 (HMGB1), an alarmin protein released from dying tumor cells, which acts as an endogenous ligand for Toll-like receptor 2 (TLR2) signaling on bone marrow-derived GBM-infiltrating DCs. METHODS AND FINDINGS Using a combined immunotherapy/conditional cytotoxic approach that utilizes adenoviral vectors (Ad) expressing Fms-like tyrosine kinase 3 ligand (Flt3L) and thymidine kinase (TK) delivered into the tumor mass, we demonstrated that CD4(+) and CD8(+) T cells were required for tumor regression and immunological memory. Increased numbers of bone marrow-derived, tumor-infiltrating myeloid DCs (mDCs) were observed in response to the therapy. Infiltration of mDCs into the GBM, clonal expansion of antitumor T cells, and induction of an effective anti-GBM immune response were TLR2 dependent. We then proceeded to identify the endogenous ligand responsible for TLR2 signaling on tumor-infiltrating mDCs. We demonstrated that HMGB1 was released from dying tumor cells, in response to Ad-TK (+ gancyclovir [GCV]) treatment. Increased levels of HMGB1 were also detected in the serum of tumor-bearing Ad-Flt3L/Ad-TK (+GCV)-treated mice. Specific activation of TLR2 signaling was induced by supernatants from Ad-TK (+GCV)-treated GBM cells; this activation was blocked by glycyrrhizin (a specific HMGB1 inhibitor) or with antibodies to HMGB1. HMGB1 was also released from melanoma, small cell lung carcinoma, and glioma cells treated with radiation or temozolomide. Administration of either glycyrrhizin or anti-HMGB1 immunoglobulins to tumor-bearing Ad-Flt3L and Ad-TK treated mice, abolished therapeutic efficacy, highlighting the critical role played by HMGB1-mediated TLR2 signaling to elicit tumor regression. Therapeutic efficacy of Ad-Flt3L and Ad-TK (+GCV) treatment was demonstrated in a second glioma model and in an intracranial melanoma model with concomitant increases in the levels of circulating HMGB1. CONCLUSIONS Our data provide evidence for the molecular and cellular mechanisms that support the rationale for the clinical implementation of antibrain cancer immunotherapies in combination with tumor killing approaches in order to elicit effective antitumor immune responses, and thus, will impact clinical neuro-oncology practice.
Collapse
Affiliation(s)
- James F Curtin
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Naiyou Liu
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Marianela Candolfi
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Weidong Xiong
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Hikmat Assi
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Kader Yagiz
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Matthew R Edwards
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
| | - Kathrin S Michelsen
- Inflammatory Bowel Disease Center and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California, United States of America
| | - Kurt M Kroeger
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Chunyan Liu
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - A. K. M. Ghulam Muhammad
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Mary C Clark
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
| | - Moshe Arditi
- Division of Pediatric Infectious Diseases, Cedars Sinai Medical Center, Los Angeles, California, United States of America
| | - Begonya Comin-Anduix
- Department of Surgery, University of California, Los Angeles, California, United States of America
| | - Antoni Ribas
- Department of Surgery, University of California, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Pedro R Lowenstein
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- The Brain Research Institute, University of California, Los Angeles, California, United States of America
| | - Maria G Castro
- Board of Governors' Gene Therapeutics Research Institute and Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- The Brain Research Institute, University of California, Los Angeles, California, United States of America
| |
Collapse
|
66
|
|