1
|
Cheng J, Chen G, Lv H, XU L, LIU H, Chen T, Qu L, Wang J, Cheng L, Hu S, Wang Y. CD4-Targeted T Cells Rapidly Induce Remissions in Mice with T Cell Lymphoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6614784. [PMID: 33855074 PMCID: PMC8019637 DOI: 10.1155/2021/6614784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 03/05/2021] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To explore the immune cell therapy for T cell lymphoma, we developed CD4-specific chimeric antigen receptor- (CAR-) engineered T cells (CD4CART), and the cytotoxic effects of CD4CART cells were determined in vitro and in vivo. METHODS CD4CART cells were obtained by transduction of lentiviral vector encoding a single-chain antibody fragment (scFv) specific for CD4 antigen, costimulatory factor CD28 fragment, and intracellular signal transduction domain of CD3 fragments. Control T cells were obtained by transduction of reporter lentiviral vector. The cytotoxicity, tumor growth, and survival rate of mice with T cell lymphoma were analyzed after adoptive T cell transfer in vivo. RESULTS CD4CART cells had potent cytotoxic activity against CD4+ T1301 tumor T cells in a concentration-dependent manner. In addition, adoptive CD4CART cell transfer significantly suppressed tumor growth and improved animal survival with T cell lymphoma, compared to the mice who received control T cells and PBS. CONCLUSION CD4CART cells have potent cytotoxic effects on T cell lymphoma. The study provided an experimental basis for CD4CART-mediated therapy of T cell lymphoma.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Hematology, Anhui Provincial Children's Hospital, Anhui Province, China
| | - Guanghua Chen
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Jiangsu Province, China
| | - Hui Lv
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Liangjing XU
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Jiangsu Province, China
| | - Huiwen LIU
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Jiangsu Province, China
| | - Tianping Chen
- Department of Hematology, Anhui Provincial Children's Hospital, Anhui Province, China
| | - Lijun Qu
- Department of Hematology, Anhui Provincial Children's Hospital, Anhui Province, China
| | - Jian Wang
- Department of Hematology, Anhui Provincial Children's Hospital, Anhui Province, China
| | - Lemei Cheng
- Department of Hematology, Anhui Provincial Children's Hospital, Anhui Province, China
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yi Wang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Patasic L, Seifried J, Bezler V, Kaljanac M, Schneider IC, Schmitz H, Tondera C, Hartmann J, Hombach A, Buchholz CJ, Abken H, König R, Cichutek K. Designed Ankyrin Repeat Protein (DARPin) to target chimeric antigen receptor (CAR)-redirected T cells towards CD4 + T cells to reduce the latent HIV + cell reservoir. Med Microbiol Immunol 2020; 209:681-691. [PMID: 32918599 PMCID: PMC7568711 DOI: 10.1007/s00430-020-00692-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 08/19/2020] [Indexed: 10/25/2022]
Abstract
Chimeric Antigen Receptor (CAR)-redirected T cells show great efficacy in the patient-specific therapy of hematologic malignancies. Here, we demonstrate that a DARPin with specificity for CD4 specifically redirects and triggers the activation of CAR engineered T cells resulting in the depletion of CD4+ target cells aiming for elimination of the human immunodeficiency virus (HIV) reservoir.
Collapse
Affiliation(s)
- Lea Patasic
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | - Janna Seifried
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany.
- Department for Infectious Disease Epidemiology, Robert Koch-Institute, Berlin, Germany.
| | - Valerie Bezler
- Regensburg Center for Interventional Immunology (RCI), Department of Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Marcell Kaljanac
- Regensburg Center for Interventional Immunology (RCI), Department of Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Irene C Schneider
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Heike Schmitz
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Jessica Hartmann
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Andreas Hombach
- Center for Molecular Medicine Cologne, University of Cologne, and Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne, University of Cologne, and Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
- Regensburg Center for Interventional Immunology (RCI), Department of Genetic Immunotherapy, University Hospital Regensburg, Regensburg, Germany
| | - Renate König
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany
- German Center for Infection Research (DZIF), Langen, Germany
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Klaus Cichutek
- Host-Pathogen Interactions, Paul-Ehrlich-Institut, Langen, Germany.
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany.
- German Center for Infection Research (DZIF), Langen, Germany.
| |
Collapse
|
3
|
Pinz KG, Yakaboski E, Jares A, Liu H, Firor AE, Chen KH, Wada M, Salman H, Tse W, Hagag N, Lan F, Leung ELH, Jiang X, Ma Y. Targeting T-cell malignancies using anti-CD4 CAR NK-92 cells. Oncotarget 2017; 8:112783-112796. [PMID: 29348865 PMCID: PMC5762550 DOI: 10.18632/oncotarget.22626] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) are a group of very aggressive non-Hodgkin's lymphomas (NHLs) with poor prognoses and account for a majority of T-cell malignancies. Overall, the standard of care for patients with T-cell malignancies is poorly established, and there is an urgent clinical need for a new approach. As demonstrated in B-cell malignancies, chimeric antigen receptor (CAR) immunotherapy provides great hope as a curative treatment regimen. Because PTCLs develop from mature T-cells, these NHLs are commonly CD4+, and CD4 is highly and uniformly expressed. Therefore, CD4 is an ideal target for PTCL CAR immunotherapy. To that effect, we created a robust third-generation anti-CD4 CAR construct (CD4CAR) and introduced it into clonal NK cells (NK-92). CD4CAR NK-92 cells specifically and robustly eliminated diverse CD4+ human T-cell leukemia and lymphoma cell lines (KARPAS-299, CCRF-CEM, and HL60) and patient samples ex vivo. Furthermore, CD4CAR NK-92 cells effectively targeted KARPAS-299 cells in vivo that modeled difficult-to-access lymphoma nodules, significantly prolonging survival. In our study, we present novel targeting of CD4 using CAR-modified NK cells, and demonstrate efficacy. Combined, our data support CD4CAR NK cell immunotherapy as a potential new avenue for the treatment of PTCLs and CD4+ T-cell malignancies.
Collapse
Affiliation(s)
- Kevin G. Pinz
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Elizabeth Yakaboski
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Alexander Jares
- Department of Pathology, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Hua Liu
- Department of Pathology, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Amelia E. Firor
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Kevin H. Chen
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Masayuki Wada
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Huda Salman
- Department of Internal Medicine, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - William Tse
- Division of Hematology and Medical Oncology, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Nabil Hagag
- Department of Internal Medicine, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Fengshuo Lan
- Department of Internal Medicine, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau SAR, China
| | - Xun Jiang
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau SAR, China
| | - Yupo Ma
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau SAR, China
- Department of Pathology, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| |
Collapse
|
4
|
Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells. Leukemia 2015; 30:701-7. [PMID: 26526988 DOI: 10.1038/leu.2015.311] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022]
Abstract
Peripheral T-cell lymphomas (PTCLs) are aggressive lymphomas with no effective upfront standard treatment and ineffective options in relapsed disease, resulting in poorer clinical outcomes as compared with B-cell lymphomas. The adoptive transfer of T cells engineered to express chimeric antigen receptors (CARs) is a promising new approach for treatment of hematological malignancies. However, preclinical reports of targeting T-cell lymphoma with CARs are almost non-existent. Here we have designed a CAR, CD4CAR, which redirects the antigen specificity of CD8+ cytotoxic T cells to CD4-expressing cells. CD4CAR T cells derived from human peripheral blood mononuclear cells and cord blood effectively redirected T-cell specificity against CD4+ cells in vitro. CD4CAR T cells efficiently eliminated a CD4+ leukemic cell line and primary CD4+ PTCL patient samples in co-culture assays. Notably, CD4CAR T cells maintained a central memory stem cell-like phenotype (CD8+CD45RO+CD62L+) under standard culture conditions. Furthermore, in aggressive orthotropic T-cell lymphoma models, CD4CAR T cells efficiently suppressed the growth of lymphoma cells while also significantly prolonging mouse survival. Combined, these studies demonstrate that CD4CAR-expressing CD8+ T cells are efficacious in ablating malignant CD4+ populations, with potential use as a bridge to transplant or stand-alone therapy for the treatment of PTCLs.
Collapse
|
5
|
The size of the viral inoculum contributes to the outcome of hepatitis B virus infection. J Virol 2009; 83:9652-62. [PMID: 19625407 DOI: 10.1128/jvi.00867-09] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The impact of virus dose on the outcome of infection is poorly understood. In this study we show that, for hepatitis B virus (HBV), the size of the inoculum contributes to the kinetics of viral spread and immunological priming, which then determine the outcome of infection. Adult chimpanzees were infected with a serially diluted monoclonal HBV inoculum. Unexpectedly, despite vastly different viral kinetics, both high-dose inocula (10(10) genome equivalents [GE] per animal) and low-dose inocula (10 degrees GE per animal) primed the CD4 T-cell response after logarithmic spread was detectable, allowing infection of 100% of hepatocytes and requiring prolonged immunopathology before clearance occurred. In contrast, intermediate (10(7) and 10(4) GE) inocula primed the T-cell response before detectable logarithmic spread and were abruptly terminated with minimal immunopathology before 0.1% of hepatocytes were infected. Surprisingly, a dosage of 10(1) GE primed the T-cell response after all hepatocytes were infected and caused either prolonged or persistent infection with severe immunopathology. Finally, CD4 T-cell depletion before inoculation of a normally rapidly controlled inoculum precluded T-cell priming and caused persistent infection with minimal immunopathology. These results suggest that the relationship between the kinetics of viral spread and CD4 T-cell priming determines the outcome of HBV infection.
Collapse
|
6
|
Grakoui A, Shoukry NH, Woollard DJ, Han JH, Hanson HL, Ghrayeb J, Murthy KK, Rice CM, Walker CM. HCV persistence and immune evasion in the absence of memory T cell help. Science 2003; 302:659-62. [PMID: 14576438 DOI: 10.1126/science.1088774] [Citation(s) in RCA: 614] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spontaneous resolution of hepatitis C virus (HCV) infection in humans usually affords long-term immunity to persistent viremia and associated liver diseases. Here, we report that memory CD4+ Tcells are essential for this protection. Antibody-mediated depletion of CD4+ Tcells before reinfection of two immune chimpanzees resulted in persistent, low-level viremia despite functional intra-hepatic memory CD8+ Tcell responses. Incomplete control of HCV replication by memory CD8+ Tcells in the absence of adequate CD4+ Tcell help was associated with emergence of viral escape mutations in class I major histocompatibility complex-restricted epitopes and failure to resolve HCV infection.
Collapse
Affiliation(s)
- Arash Grakoui
- Center for the Study of Hepatitis C, Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Shen G, Zhu H, Wang X, Zhang Y, Zhu Z, Wang S. Anti-proliferative effects induced by anti-CD4 human/murine chimeric antibody and murine anti-CD4 monoclonal antibody. JOURNAL OF TONGJI MEDICAL UNIVERSITY = TONG JI YI KE DA XUE XUE BAO 2003; 19:6-9. [PMID: 12840865 DOI: 10.1007/bf02895584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The effects of chimeric anti-CD4 human/murine chimeric antibody and murine anti-CD4 monoclonal antibody (McAb) on the proliferation induced by anti-CD3 McAb, phytohemagglutinin (PHA), IL-2, and allogeneic cells were studied. The results showed that chimeric anti-CD4 antibody and murine anti-CD4 McAb could inhibit the proliferation induced by the above inducers and the inhibitory effects were related to the dosage of the antibodies.
Collapse
Affiliation(s)
- G Shen
- Department of Immunology, Tongji Medical University, Wuhan 430030
| | | | | | | | | | | |
Collapse
|
8
|
Thimme R, Wieland S, Steiger C, Ghrayeb J, Reimann KA, Purcell RH, Chisari FV. CD8(+) T cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infection. J Virol 2003; 77:68-76. [PMID: 12477811 PMCID: PMC140637 DOI: 10.1128/jvi.77.1.68-76.2003] [Citation(s) in RCA: 727] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Although the CD4(+)- and CD8(+)-T-cell responses to the hepatitis B virus (HBV) are thought to be crucial for the control of HBV infection, the relative contribution of each T-cell subset as an effector of viral clearance is not known. To examine this question, we monitored the course of HBV infection in control, CD4-depleted, and CD8-depleted chimpanzees. Our results demonstrate that CD8(+) cells are the main effector cells responsible for viral clearance and disease pathogenesis during acute HBV infection, and they suggest that viral clearance is mediated by both noncytolytic and cytolytic effector functions of the CD8(+)-T-cell response.
Collapse
Affiliation(s)
- Robert Thimme
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Fishwild DM, Hudson DV, Deshpande U, Kung AH. Differential effects of administration of a human anti-CD4 monoclonal antibody, HM6G, in nonhuman primates. Clin Immunol 1999; 92:138-52. [PMID: 10444358 DOI: 10.1006/clim.1999.4734] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A human sequence IgGkappa anti-CD4 monoclonal antibody (mAb), HM6G, originally isolated from a human immunoglobulin transgenic mouse was specific for and bound with high binding avidity to the CD4 antigen expressed on human, chimpanzee, and cynomolgus monkey T cells. Prior to testing this mAb in human clinical trials, a number of preclinical primate studies were performed. In chimpanzees, HM6G did not deplete circulating CD4(+) T cells and was cleared in a dose-dependent manner. In contrast, this mAb administered to cynomolgus monkeys depleted CD4(+) T cells (albeit only at high doses) and its clearance, which had reached saturation even at very low doses, was much slower. These differences were most likely due to the additional and rather substantial expression of the CD4 antigen on chimpanzee monocytes. In monkeys, the T cell depletion was mitigated by infusing the mAb over 30 min or longer (as opposed to 30 s) while only slightly altering the clearance. As expected, the human mAb did not induce an immune response in chimpanzees, although it did induce a low titer response in monkeys. These disparate pharmacokinetic and pharmacodynamic results suggest prudence when extrapolating results obtained in nonhuman models to humans.
Collapse
Affiliation(s)
- D M Fishwild
- GenPharm International, San Jose, California, 95131, USA.
| | | | | | | |
Collapse
|
10
|
Naessens J, Scheerlinck JP, De Buysscher EV, Kennedy D, Sileghem M. Effective in vivo depletion of T cell subpopulations and loss of memory cells in cattle using mouse monoclonal antibodies. Vet Immunol Immunopathol 1998; 64:219-34. [PMID: 9730218 DOI: 10.1016/s0165-2427(98)00138-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Conditions were established to obtain depletion of T lymphocyte subsets in lymphoid tissues of calves by injection of mouse monoclonal antibodies to T cell antigens. Adverse reactions were avoided by injecting small quantities of antibody, until target cells had disappeared from blood. Two different mechanisms appeared to be responsible for elimination of the target cells. Rapid depletion of T cells was associated with complement-binding antibody isotypes (IgG2a, IgM), suggesting a complement-mediated mechanism. Clearance of T cells after several days was observed with a non complement-binding isotype (IgG1), suggesting phagocytosis or induction of apoptosis as possible mechanisms. Clearance of the cells in peripheral blood and spleen was obtained with 10-20 mg of anti-CD4 or anti-CD8, but almost ten times as much was needed to obtain depletion of the cells in lymph nodes and Peyer's patches. Depletion lasted for 12 days for CD4 T cells and 3 weeks for CD8 T cells. Successful and lasting depletion (at least 2 weeks) was also obtained with other T cell reagents, such as anti-CD2 and anti-WC1 (gamma/delta T cells). Although B lymphocytes could be removed by a complement-binding antibody, complete depletion of these cells only lasted for a few hours, probably because B cells regenerate faster than T cells. T cell function was severely inhibited when CD4+ T cells were depleted. Stimulation of T cells with foot and mouth disease viral antigen (FMDV) in vaccinated calves was non-existent after depletion. Even 2 months after restoration of normal CD4 T cell levels in blood, activity to FMDV was low. This suggested that the depleted T cells were replaced by naive cells.
Collapse
Affiliation(s)
- J Naessens
- International Livestock Research Institute, Nairobi, Kenya.
| | | | | | | | | |
Collapse
|
11
|
Reimann KA, Lin W, Bixler S, Browning B, Ehrenfels BN, Lucci J, Miatkowski K, Olson D, Parish TH, Rosa MD, Oleson FB, Hsu YM, Padlan EA, Letvin NL, Burkly LC. A humanized form of a CD4-specific monoclonal antibody exhibits decreased antigenicity and prolonged plasma half-life in rhesus monkeys while retaining its unique biological and antiviral properties. AIDS Res Hum Retroviruses 1997; 13:933-43. [PMID: 9223409 DOI: 10.1089/aid.1997.13.933] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Certain monoclonal antibodies (MAbs) directed against CD4 can efficiently block HIV-1 replication in vitro. To explore CD4-directed passive immunotherapy for prevention or treatment of AIDS virus infection, we previously examined the biological activity of a nondepleting CD4-specific murine MAb, mu5A8. This MAb, specific for domain 2 of CD4, blocks HIV-1 replication at a post-gp120-CD4 binding step. When administered to normal rhesus monkeys, all CD4+ target cells were coated with antibody, yet no cell clearance or measurable immunosuppression occurred. However, strong anti-mouse Ig responses rapidly developed in all monkeys. In the present study, we report a successfully humanized form of mu5A8 (hu5A8) that retains binding to both human and monkey CD4 and anti-AIDS virus activity. When administered intravenously to normal rhesus monkeys, hu5A8 bound to all target CD4+ cells without depletion and showed a significantly longer plasma half-life than mu5A8. Nevertheless, an anti-hu5A8 response directed predominantly against V region determinants did eventually appear within 2 to 4 weeks in most animals. However, when hu5A8 was administered to rhesus monkeys chronically infected with the simian immunodeficiency virus of macaques, anti-hu5A8 antibodies were not detected. Repeated administration of hu5A8 in these animals resulted in sustained plasma levels and CD4+ cell coating with humanized antibody for 6 weeks. These studies demonstrate the feasibility of chronic administration of CD4-specific MAb as a potential means of treating or preventing HIV-1 infection.
Collapse
Affiliation(s)
- K A Reimann
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Anderson D, Chambers K, Hanna N, Leonard J, Reff M, Newman R, Baldoni J, Dunleavy D, Reddy M, Sweet R, Truneh A. A primatized MAb to human CD4 causes receptor modulation, without marked reduction in CD4+ T cells in chimpanzees: in vitro and in vivo characterization of a MAb (IDEC-CE9.1) to human CD4. CLINICAL IMMUNOLOGY AND IMMUNOPATHOLOGY 1997; 84:73-84. [PMID: 9191886 DOI: 10.1006/clin.1997.4363] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A Primatized anti-CD4 monoclonal antibody (MAb), CE9.1, with V-domain from cynomolgus macaque (showing 92% homology with human consensus sequence V-domains), and a human IgG1 constant region, was characterized in vitro and in vivo in chimpanzees. This MAb binds human CD4 with Kd of 1.0 nM and was also able to bind to human IgG Fc receptors (Fc gamma R). However, despite being of the IgG1 subclass, CE9.1 did not bind to complement component C1q, nor did it mediate complement-dependent cytotoxicity. Examination of T cells from a number of species showed restricted reactivity for CE9.1, recognizing only human and chimpanzee CD4. In both human and chimpanzee MLRs, it had an IC50 of about 10.0 ng/mL. Therefore, a chimpanzee in vivo model was used to characterize CE9.1, CE9.1 caused transient decrease in the number of lymphocytes bearing the CD4 receptor starting at doses of 0.3 mg/kg in an in vivo dose ranging study in one chimpanzee. This effect was reversed within approximately 7 days. In a multiple high-dose study in which 10.0 mg/kg of CE9.1 was administered at intervals of 1-3 months, there was a dramatic loss of CD4 marker with a reciprocal increase in the number of CD3+ CD8- CD4- cells. The CD4 receptor was totally undetectable on these lymphocytes for 1-2 weeks, with a gradual, but complete, reversal within 4 weeks. We interpret these observations as receptor modulation because, although there was apparent loss of CD4+ lymphocytes, an equivalent number of CD3+CD8- T lymphocytes were present in circulation in all four chimpanzees treated with 10.0 mg/kg CE9.1. Even at this high dose, only limited reduction of CD4+ T lymphocytes was observed in these animals. These observations are in sharp contrast to what has been reported in rodents or in human clinical studies using other IgG1 mAbs to human CD4. CD8 counts, although variable, remained unaffected by CE9.1 treatment. No adverse events were observed following administration of CE9.1 to chimpanzees, and there was no detectable host immune responses to the Primatized MAb.
Collapse
Affiliation(s)
- D Anderson
- IDEC Pharmaceuticals Corporation, San Diego, California 92121, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Saitovitch D, Bushell A, Mabbs DW, Morris PJ, Wood KJ. Kinetics of induction of transplantation tolerance with a nondepleting anti-Cd4 monoclonal antibody and donor-specific transfusion before transplantation. A critical period of time is required for development of immunological unresponsiveness. Transplantation 1996; 61:1642-7. [PMID: 8669111 DOI: 10.1097/00007890-199606150-00016] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The combination of a depleting anti-Cd43 monoclonal antibody (mAb) and a single donor-specific transfusion before transplantation has been shown to induce operational transplantation tolerance in the majority of cardiac allograft recipients in a mouse model. To examine a protocol which might be more clinically relevant, we have modified this tolerance-inducing protocol by substituting the depleting with a nondepleting anti-Cd4 mAb. We show that this form of pretreatment can also induce immunologic unresponsiveness in most recipients (C3H/He, H2(k)), provided a critical period of time, in this case 28 days, is allowed between pretreatment and transplantation of a fully mismatched heart graft (H2(b)). When only 1 or 2 weeks were allowed between pretreatment and transplantation, only slight graft prolongation was obtained when compared with recipients receiving anti-Cd4 mAb alone, at these time points. Maintenance of tolerance in this model was due, at least in part, to active mechanisms as immunologic unresponsiveness to donor antigens could be transferred to naive syngeneic mice by splenocytes from recipients bearing long-term functioning grafts. These findings suggest that a population of regulatory cells develop after pretreatment with nondepleting anti-Cd4 mAb and donor-specific transfusion, and that it takes at least 1 month for these cells to expand and effectively drive the recipient's immune system toward immunologic unresponsiveness.
Collapse
Affiliation(s)
- D Saitovitch
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Headington, Oxford, England
| | | | | | | | | |
Collapse
|
14
|
Chirmule N, Pahwa S. Envelope glycoproteins of human immunodeficiency virus type 1: profound influences on immune functions. Microbiol Rev 1996; 60:386-406. [PMID: 8801439 PMCID: PMC239449 DOI: 10.1128/mr.60.2.386-406.1996] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Infection by human immunodeficiency virus type 1 (HIV-1) leads to progressive destruction of the CD4+ T-cell subset, resulting in immune deficiency and AIDS. The specific binding of the viral external envelope glycoprotein of HIV-1, gp120, to the CD4 molecules initiates viral entry. In the past few years, several studies have indicated that the interaction of HIV-1 envelope glycoprotein with cells and molecules of the immune system leads to pleiotropic biological effects on immune functions, which include effects on differentiation of CD34+ lymphoid progenitor cells and thymocytes, aberrant activation and cytokine secretion patterns of mature T cells, induction of apoptosis, B-cell hyperactivity, inhibition of T-cell dependent B-cell differentiation, modulation of macrophage functions, interactions with components of complement, and effects on neuronal cells. The amino acid sequence homologies of the envelope glycoproteins with several cellular proteins have suggested that molecular mimicry may play a role in the pathogenesis of the disease. This review summarizes work done by several investigators demonstrating the profound biological effects of envelope glycoproteins of HIV-1 on immune system cells. Extensive studies have also been done on interactions of the viral envelope proteins with components of the immune system which may be important for eliciting a "protective immune response." Understanding the influences of HIV-1 envelope glycoproteins on the immune system may provide valuable insights into HIV-1 disease pathogenesis and carries implications for the trials of HIV-1 envelope protein vaccines and immunotherapeutics.
Collapse
Affiliation(s)
- N Chirmule
- Department of Pediatrics, North Shore University Hospital-Cornell University Medical College, Manhasset, New York 11030, USA. N_Chirmule or
| | | |
Collapse
|
15
|
t Hart BA, Otten HG. Prospects of immunotherapy for rheumatoid arthritis. PHARMACY WORLD & SCIENCE : PWS 1995; 17:178-85. [PMID: 8597773 DOI: 10.1007/bf01870608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The main challenge in the development of new modalities for the treatment of rheumatoid arthritis is to enhance the specificity while reducing the adverse side-effects of therapeutics. Biotechnology provides a variety of reagents, such as monoclonal antibodies, recombinant cytokines, cytokine antagonists, and small peptides, with the potential to interfere with selected stages of the disease process in a highly specific manner. In addition, several new therapeutic approaches have emerged as a result of extensive research with animal models of disease, including T-cell vaccination and bone marrow transplantation. This article discusses current insights into the pathogenesis of rheumatic diseases, focusing on rheumatoid arthritis. A number of new therapeutic modalities for rheumatoid arthritis, in particular those acting on the immune system, are discussed. Because it is not possible to provide a complete overview of all the developments in the field in limited space, a selection of strategies and modalities which are representative of the broad variety of immunotherapeutic approaches currently used are highlighted.
Collapse
Affiliation(s)
- B A t Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | | |
Collapse
|