1
|
Turaj AH, Hussain K, Cox KL, Rose-Zerilli MJJ, Testa J, Dahal LN, Chan HTC, James S, Field VL, Carter MJ, Kim HJ, West JJ, Thomas LJ, He LZ, Keler T, Johnson PWM, Al-Shamkhani A, Thirdborough SM, Beers SA, Cragg MS, Glennie MJ, Lim SH. Antibody Tumor Targeting Is Enhanced by CD27 Agonists through Myeloid Recruitment. Cancer Cell 2017; 32:777-791.e6. [PMID: 29198913 PMCID: PMC5734932 DOI: 10.1016/j.ccell.2017.11.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/28/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) can destroy tumors by recruiting effectors such as myeloid cells, or targeting immunomodulatory receptors to promote cytotoxic T cell responses. Here, we examined the therapeutic potential of combining a direct tumor-targeting mAb, anti-CD20, with an extended panel of immunomodulatory mAbs. Only the anti-CD27/CD20 combination provided cures. This was apparent in multiple lymphoma models, including huCD27 transgenic mice using the anti-huCD27, varlilumab. Detailed mechanistic analysis using single-cell RNA sequencing demonstrated that anti-CD27 stimulated CD8+ T and natural killer cells to release myeloid chemo-attractants and interferon gamma, to elicit myeloid infiltration and macrophage activation. This study demonstrates the therapeutic advantage of using an immunomodulatory mAb to regulate lymphoid cells, which then recruit and activate myeloid cells for enhanced killing of mAb-opsonized tumors.
Collapse
Affiliation(s)
- Anna H Turaj
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Khiyam Hussain
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Kerry L Cox
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Matthew J J Rose-Zerilli
- Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - James Testa
- Celldex Therapeutics, Inc., Hampton, NJ 08827, USA
| | - Lekh N Dahal
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - H T Claude Chan
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Sonya James
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Vikki L Field
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Matthew J Carter
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Hyung J Kim
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Jonathan J West
- Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | | | - Li-Zhen He
- Celldex Therapeutics, Inc., Hampton, NJ 08827, USA
| | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, NJ 08827, USA
| | - Peter W M Johnson
- Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Aymen Al-Shamkhani
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Stephen M Thirdborough
- Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Stephen A Beers
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Martin J Glennie
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Sean H Lim
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; Cancer Research UK Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
2
|
Williams EL, Tutt AL, Beers SA, French RR, Chan CHT, Cox KL, Roghanian A, Penfold CA, Butts CL, Boross P, Verbeek JS, Cragg MS, Glennie MJ. Immunotherapy Targeting Inhibitory Fcγ Receptor IIB (CD32b) in the Mouse Is Limited by Monoclonal Antibody Consumption and Receptor Internalization. THE JOURNAL OF IMMUNOLOGY 2013; 191:4130-40. [DOI: 10.4049/jimmunol.1301430] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
3
|
Du Y, Honeychurch J, Cragg MS, Bayne M, Glennie MJ, Johnson PWM, Illidge TM. Antibody-induced intracellular signaling works in combination with radiation to eradicate lymphoma in radioimmunotherapy. Blood 2004; 103:1485-94. [PMID: 14576070 DOI: 10.1182/blood-2003-06-2037] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Radioimmunotherapy (RIT) has emerged as an effective treatment for lymphoma, however the underlying mechanisms are poorly understood. We therefore investigated the relative contributions of antibody and targeted radiation to the clearance of tumor in vivo, using 2 different syngeneic murine B-cell lymphoma models. Although RIT with 131I–anti–major histocompatibility complex class II (MHCII) was effective in targeting radiation to tumor, no improvement in survival was seen by escalating the radiation dose alone and there were no long-term survivors. In contrast, using the combination of 131I anti-MHCII in the presence of unlabeled anti-idiotype (anti-Id), 100% prolonged disease-free survival was seen in both B-cell lymphoma models at the higher radiation dose. Using in vivo tracking we show that treatment with radiation plus anti-Id monoclonal antibody (mAb) results in a substantially greater reduction of splenic tumor cells than with either treatment alone. Prolonged survival could also be achieved using 131I anti-MHCII plus the signaling anti-CD19 mAb. Furthermore, the ability of these anti–B-cell mAbs to improve survival with targeted radiotherapy appeared to correlate with their ability to initiate intracellular signal transduction. Together these data illustrate that using 1 mAb to target radiation to tumor and a second to induce cell signaling is an effective new strategy in RIT.
Collapse
Affiliation(s)
- Yong Du
- Cancer Sciences Division, School of Medicine, Southampton University Hospital, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
4
|
Honeychurch J, Glennie MJ, Johnson PWM, Illidge TM. Anti-CD40 monoclonal antibody therapy in combination with irradiation results in a CD8 T-cell-dependent immunity to B-cell lymphoma. Blood 2003; 102:1449-57. [PMID: 12714523 DOI: 10.1182/blood-2002-12-3717] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanisms of interaction between anti-CD40 monoclonal antibody (mAb) therapy and external beam irradiation were investigated in 2 syngeneic B-cell lymphoma models. We have established doses of anti-CD40 mAb and irradiation which, although ineffective when given singly, were capable of providing long-term protection when used in combination. Furthermore, such treatment was not only critically dependent on the dose of mAb and irradiation but also on tumor load, with greater efficacy only occurring at higher tumor burden. Using blocking antibody, the potency of treatment was shown to be totally dependent on CD8+ T cells, with protective levels of CD8+ cells occurring only in mice receiving the combination of anti-CD40 and irradiation. Interestingly, the ratio of T cells (CD8+) to tumor cells in mice receiving combination treatment was between 10 and 15 times that seen in animals given anti-CD40 or irradiation alone. In vivo tracking experiments revealed a 2-phase decrease in tumor burden, the first resulting directly from the external irradiation and the second, occurring 5 days later, concomitant with the rise in tumor-specific CD8+ cells. We suggest that the external irradiation induced an initial kill of lymphoma cells, probably by apoptosis, which releases tumor antigens and slows the progression of the malignancy to allow generation of a curative cytotoxic T lymphocyte (CTL) response promoted by the anti-CD40 mAb. Combining irradiation with immunomodulatory mAb as described here appears to provide a powerful new approach to the management of cancer.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Apoptosis/immunology
- Apoptosis/radiation effects
- CD40 Antigens/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/radiation effects
- Combined Modality Therapy
- Cytotoxicity, Immunologic/immunology
- Disease Models, Animal
- Dose-Response Relationship, Radiation
- Immunization, Passive/methods
- Immunotherapy, Adoptive
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/radiotherapy
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Tumor Cells, Cultured
- Whole-Body Irradiation
Collapse
Affiliation(s)
- Jamie Honeychurch
- Cancer Research UK Oncology Unit, Tenovus Research Laboratory, Cancer Sciences Division, School of Medicine, Southampton General Hospital, United Kingdom
| | | | | | | |
Collapse
|
5
|
Tutt AL, O'Brien L, Hussain A, Crowther GR, French RR, Glennie MJ. T cell immunity to lymphoma following treatment with anti-CD40 monoclonal antibody. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:2720-8. [PMID: 11884438 DOI: 10.4049/jimmunol.168.6.2720] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this study we demonstrate that treatment with anti-CD40 mAb eradicates a range of mouse lymphomas (BCL(1), A31, A20, and EL4), but only when used against i.v. tumor doses in excess of 10(7) cells. Only partial protection was seen against smaller tumor loads. We saw no evidence that anti-CD40 mAb changed the phenotype of the lymphomas or inhibited their growth in the initial period following treatment, but it did result in a rapid expansion of cytotoxic CD8(+) cells that was able to clear the neoplastic disease and provide long-term protection against tumor rechallenge. The CTL responses were blocked by mAb against a range of coreceptors and cytokines, including CD8, B7-1, B7-2, LFA-1, and IFN-gamma, but not CD4 or CTLA-4, indicating the presence of a conventional cellular Th1 response. Furthermore, we found evidence of cross-recognition between lymphomas (BCL(1) and A20) as measured by cytotoxicity and IFN-gamma responses in vitro and using tumor rechallenge experiments, suggesting common target Ags. Finally, although anti-CD40 was shown to stimulate NK cell killing, we could find no role for these cells in controlling tumor growth. These data underline the ability of anti-CD40 mAb to potentiate CTL responses and the potency of cellular immunity in eradicating large quantities of syngeneic tumor.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/therapeutic use
- CD40 Antigens/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Division/immunology
- Cytotoxicity, Immunologic/immunology
- Dose-Response Relationship, Immunologic
- Immunity, Innate
- Injections, Intravenous
- Killer Cells, Natural/immunology
- Lymphocyte Activation/immunology
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Neoplasm Transplantation
- T-Lymphocytes/immunology
- Thymoma/immunology
- Thymoma/pathology
- Thymoma/prevention & control
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Alison L Tutt
- Tenovus Research Laboratory, Cancer Sciences Division, University School of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | | | | | | | | | | |
Collapse
|
6
|
Abstract
CD64 (FcγRI) receptors represent highly potent trigger molecules for activated polymorphonuclear cells (PMN) and mediate lysis of a range of tumors in the presence of appropriate monoclonal antibodies. An huCD64 transgenic mouse model designed to analyze the therapeutic activity of a panel of bispecific F(ab')2(BsAb) in retargeting granulocyte–colony-stimulating factor (G-CSF)–activated PMN against syngeneic B-cell lymphomas is reported. This model allows careful analysis of the individual elements of the therapeutic process. BsAb were directed against immunoglobulin-idiotype (Id), major histocompatibility class II (MHC II), or CD19 on the tumors and huCD64 on the effectors. In vitro cytotoxicity assays and in vivo tumor tracking showed that, provided effectors were activated with G-CSF, all 3 derivatives destroyed and cleared lymphoma cells, with (huCD64 × MHC II) proving by far the most cytotoxic in vitro. However, though all derivatives delivered some survival advantage, only the [huCD64 × Id] BsAb provided long-term protection to tumor-bearing animals. These results demonstrate that CD64-recruited cytotoxic effectors operate in vivo but that the (huCD64 × Id) conferred an additional anti-tumor function essential for long-term protection. T-cell depletion studies demonstrated that this extra therapeutic activity with [huCD64 × Id] was totally dependent on CD4 and CD8 T cells and that mice, once “cured” with BsAb, were resistant to tumor rechallenge. These findings indicate that CD64 is an effective trigger molecule for delivering cytokine-activated PMN against tumor in vivo and that, provided tumor targets are selected appropriately, CD64-based BsAb can establish long-term T-cell immunity.
Collapse
|
7
|
Abstract
Abstract
CD64 (FcγRI) receptors represent highly potent trigger molecules for activated polymorphonuclear cells (PMN) and mediate lysis of a range of tumors in the presence of appropriate monoclonal antibodies. An huCD64 transgenic mouse model designed to analyze the therapeutic activity of a panel of bispecific F(ab')2(BsAb) in retargeting granulocyte–colony-stimulating factor (G-CSF)–activated PMN against syngeneic B-cell lymphomas is reported. This model allows careful analysis of the individual elements of the therapeutic process. BsAb were directed against immunoglobulin-idiotype (Id), major histocompatibility class II (MHC II), or CD19 on the tumors and huCD64 on the effectors. In vitro cytotoxicity assays and in vivo tumor tracking showed that, provided effectors were activated with G-CSF, all 3 derivatives destroyed and cleared lymphoma cells, with (huCD64 × MHC II) proving by far the most cytotoxic in vitro. However, though all derivatives delivered some survival advantage, only the [huCD64 × Id] BsAb provided long-term protection to tumor-bearing animals. These results demonstrate that CD64-recruited cytotoxic effectors operate in vivo but that the (huCD64 × Id) conferred an additional anti-tumor function essential for long-term protection. T-cell depletion studies demonstrated that this extra therapeutic activity with [huCD64 × Id] was totally dependent on CD4 and CD8 T cells and that mice, once “cured” with BsAb, were resistant to tumor rechallenge. These findings indicate that CD64 is an effective trigger molecule for delivering cytokine-activated PMN against tumor in vivo and that, provided tumor targets are selected appropriately, CD64-based BsAb can establish long-term T-cell immunity.
Collapse
|
8
|
Tutt AL, French RR, Illidge TM, Honeychurch J, McBride HM, Penfold CA, Fearon DT, Parkhouse RME, Klaus GGB, Glennie MJ. Monoclonal Antibody Therapy of B Cell Lymphoma: Signaling Activity on Tumor Cells Appears More Important Than Recruitment of Effectors. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.6.3176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Despite the recent success of mAb in the treatment of certain malignancies, there is still considerable uncertainty about the mechanism of action of anti-cancer Abs. Here, a panel of rat anti-mouse B cell mAb, including Ab directed at surface IgM Id, CD19, CD22, CD40, CD74, and MHC class II, has been investigated in the treatment of two syngeneic mouse B cell lymphomas, BCL1 and A31. Only three mAb were therapeutically active in vivo, anti-Id, anti-CD19, and anti-CD40. mAb to the other Ags showed little or no therapeutic activity in either model despite giving good levels of surface binding and activity in Ag-dependent cellular cytotoxicity and complement assays, and in some cases inhibiting cell growth in vitro. We conclude that the activity of mAb in vitro does not predict therapeutic performance in vivo. Furthermore, in vivo tracking experiments using fluorescently tagged cells showed that anti-Id and anti-CD40 mAb probably operate via different mechanisms: the anti-Id mAb cause growth arrest that is almost immediate and does not eliminate cells over a period of 5 or 6 days, and the anti-CD40 mAb have a delayed effect that allows tumor to grow normally for 3 days, but then abruptly eradicates lymphoma cells. This work supports the belief that mAb specificity is critical to therapeutic success in lymphoma and that, in addition to any effector-recruiting activity they may possess, in vivo mAb operate via mechanisms that involve cross-linking and signaling of key cellular receptors.
Collapse
Affiliation(s)
- Alison L. Tutt
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| | - Ruth R. French
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| | - Timothy M. Illidge
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| | - Jamie Honeychurch
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| | - Harry M. McBride
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| | - Christine A. Penfold
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| | - Douglas T. Fearon
- †Wellcome Trust Immunology Center, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | | | | - Martin J. Glennie
- *Lymphoma Research Unit, Tenovus Laboratory, General Hospital, Southampton, United Kingdom
| |
Collapse
|
9
|
Montgomery AM, Wing MG, Lachmann PJ. The targeting of T-helper cells and tumourcidal macrophages to a B-cell lymphoma using a PPD-monoclonal antibody heteroconjugate. Immunol Suppl 1992; 75:217-23. [PMID: 1532376 PMCID: PMC1384697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This paper describes a T-cell targeting strategy based on the use of an antigen-monoclonal antibody heteroconjugate. A rat anti-idiotypic monoclonal antibody specific for a murine B-cell lymphoma was conjugated to the purified protein derivative (PPD) of tuberculin. This construct selectively delivered up to 4.5 x 10(4) molecules of PPD onto each tumour cell. Targeted PPD was internalized for endosomal processing and was presented in association with the I-A class II restriction element to PPD-reactive T-helper (Th) cells. Activated Th cells were demonstrated to proliferate and secrete significant levels of tumour necrosis factor (TNF). Such lymphokine secretion was observed at a PPD concentration as low as 1 ng/ml. Despite the secretion of TNF, the B-cell lymphoma was found to be resistant to autonomous Th-mediated cytotoxicity. Targeted Th cells did, however, activate tumourcidal macrophages that subsequently mediated significant tumour cytostasis. Based on this observation, it is proposed that the targeting system described may be exploited as the basis for a future immunotherapeutic strategy.
Collapse
Affiliation(s)
- A M Montgomery
- Molecular Immunopathology Unit, MRC Centre, Cambridge, U.K
| | | | | |
Collapse
|
10
|
George AJ, Foreman RC, Stevenson FK. Characterisation of a light chain loss variant of the BCL1 lymphoma. Mol Immunol 1991; 28:789-99. [PMID: 1649967 DOI: 10.1016/0161-5890(91)90122-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Vaccination of BALB/c mice with idiotypic (id) IgM derived from the murine B cell lymphoma BCL1, protects the animals from challenge with tumour cells. Escape of the tumour cells from immune control is associated with the selection of variant cells which fail to express significant levels of id IgM on their cell surface. We have previously isolated one such variant, SNAG 1, and shown that, while it expresses less than 10% of the levels of surface IgM of the parental BCL1 lymphoma, it continues to synthesise id material which can be detected within the cell. In this report we present a detailed characterisation of this variant and show that the tumour cells no longer synthesise the lambda light chain. This failure to produce the light chain causes the mu heavy chains in SNAG 1 to remain marooned in the endoplasmic reticulum. The mu heavy chains in SNAG 1 have a normal mol. wt and isoelectric point, and so appear not to be mutated. This is unlike the vast majority of light chain loss variants, in which the heavy chains have been shown to contain deletions. Investigation of the mechanisms responsible for the loss of light chain synthesis demonstrated that, while mRNA for the light chain is present, and of a normal size, there was no production of light chain protein in a cell free system. This indicates that the failure to express light chain by SNAG 1 cells is due to an inability to translate the light chain mRNA into the detectable levels of lambda light chain protein.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Blotting, Northern
- Cell-Free System
- Cytoplasm/immunology
- DNA Probes
- Enzyme-Linked Immunosorbent Assay
- Female
- Glycosylation
- Immunoglobulin Heavy Chains/immunology
- Immunoglobulin Light Chains/genetics
- Immunoglobulin Light Chains/immunology
- Immunoglobulin M/biosynthesis
- Immunoglobulin M/metabolism
- Immunophenotyping
- Isoelectric Focusing
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Peroxidase
- Precipitin Tests
- Protein Biosynthesis
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- A J George
- Lymphoma Research Unit, Tenovus Research Laboratories, Southampton General Hospital, U.K
| | | | | |
Collapse
|
11
|
George AJ, McBride HM, Glennie MJ, Smith LJ, Stevenson FK. Monoclonal antibodies raised against the idiotype of the murine B cell lymphoma, BCL1 act primarily with heavy chain determinants. Hybridoma (Larchmt) 1991; 10:219-27. [PMID: 1908435 DOI: 10.1089/hyb.1991.10.219] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Anti-idiotypic antibodies can be used as probes to distinguish neoplastic cells from their normal counterparts. In addition they have been used in the passive therapy of B cell tumors. In this report we describe a panel of 7 rat monoclonal antibodies raised against idiotypic determinants carried by the IgM molecule of the BCL1 lymphoma. The majority (6/7) of these antibodies recognize private idiotypic determinants that are carried on the isolated mu heavy chain of the molecule, and do not require the lambda chain for reactivity. This is unusual for antibodies raised against the idiotype of the whole immunoglobulin molecule, which normally require both chains for reactivity. The antibodies do not, however, bind peptides corresponding to the complementarity determining regions of the mu heavy chain of BCL1. The antibodies perform well in complement mediated cytotoxicity, and, in at least one case, are effective in the passive immunotherapy of BCL1 lymphoma.
Collapse
MESH Headings
- Animals
- Antibodies, Anti-Idiotypic/immunology
- Antibodies, Anti-Idiotypic/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/therapeutic use
- Immunization, Passive
- Immunoglobulin Idiotypes/immunology
- Immunoglobulin mu-Chains/immunology
- Lymphoma, B-Cell/diagnosis
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Neoplasm Proteins/immunology
- Rats
Collapse
Affiliation(s)
- A J George
- Host Immunity to Tumour Group, Tenovus Research Laboratory, Southampton General Hospital, United Kingdom
| | | | | | | | | |
Collapse
|
12
|
Dyke RJ, McBride H, George AJ, Hamblin TJ, Stevenson FK. Idiotypic vaccination against B-cell lymphoma leads to dormant tumour. Cell Immunol 1991; 132:70-83. [PMID: 2065360 DOI: 10.1016/0008-8749(91)90007-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Idiotypic immunoglobulin, which can be considered to bear tumour-associated antigens in the context of B-cell lymphoma, has been obtained from the splenic A31 tumour, purified, and used to immunise syngeneic mice. On subsequent exposure to a lethal challenge of lymphoma cells, the mice showed no overt tumour development over an observation period of 6 months, whereas mice immunised with an unrelated idiotypic immunoglobulin succumbed to lymphoma after about 20 days. Anti-idiotypic immunity persisted in protected mice, since a second exposure to a lethal tumour dose 4 months after the first challenge also failed to induce lymphoma. Anti-idiotypic antibody appeared to have a major role in protection when analysed by passive transfer experiments, with no contribution from transferred cells. Protected mice were investigated for the presence of lymphoma cells 4-8 months following exposure to tumour, but the spleens, which were of normal weight and appearance, contained few or no tumour cells by phenotypic analysis. However, passage of cells dispersed from these spleens led, in 60% of cases, to tumour development in unimmunised recipients. The emergent tumours were indistinguishable from the original A31 lymphoma, with no evidence for variants, indicating that the cells were unable to grow in the immune mice, but that this dormant state could be disrupted by transfer.
Collapse
Affiliation(s)
- R J Dyke
- Lymphoma Research Unit, Tenovus Research Laboratory, General Hospital, Southampton, United Kingdom
| | | | | | | | | |
Collapse
|
13
|
George AJ, Stevenson FK. Prospects for the treatment of B cell tumors using idiotypic vaccination. Int Rev Immunol 1989; 4:271-310. [PMID: 2519930 DOI: 10.3109/08830188909044783] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Manipulation of the immune system in order specifically to attack autologous tumor cell has been a distant goal, largely due to the poor definition of tumor-associated antigens. By focussing on B cell lymphomas which express a molecularly defined clonal marker, the idiotypic immunoglobulin, it has been possible to generate autologous anti-idiotypic responses which suppress individual tumors. Studies of the components of these responses are providing insight into host effector mechanisms which can be activated against tumors, and also into the many strategies adopted by the target cells in order to avoid such attack. Promising results in various animal models where tumor bearers can be treated by idiotypic immunization have led to the point where application of this approach to the treatment of certain categories of human B cell lymphoma can be considered.
Collapse
Affiliation(s)
- A J George
- Host Immunity to Tumour Group, Southampton General Hospital, United Kingdom
| | | |
Collapse
|
14
|
Cobb LM, Butler SA. Treatment of the murine lymphoma A31 with intravenous, sterilized, 114mIn-loaded A31 cells. Radiother Oncol 1987; 10:217-30. [PMID: 3124221 DOI: 10.1016/s0167-8140(87)80008-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effect on a mouse B-cell lymphoma (A31) of intravenously (i.v.) injected, sterilized, A31 lymphoma cells loaded with [114mIn]oxine was examined. The treatment of the B-cell lymphoma was started 14 days after the i.v. injection of 10(2) viable cells. The [114mIn]oxine-bearing gamma-ray sterilized A31 cells were injected at two dose levels either with 37 kBq on days 15, 22 and 27 (total dose 110 kBq) or 74 kBq on days 15, 16, 17, 22 and 27 (total dose 370 kBq). The smaller amount (110 kBq) of 114mIn activity did not extend the survival of mice when compared with non-treatment controls but the greater amount (370 kBq) produced a significant extension in survival. This survival time was longer than that produced by 370 kBq [114mIn]oxine without carrier cells, lethal total body irradiation (with bone marrow rescue), splenectomy at day 14 or a single injection on day 14 of a maximum tolerated dose of vincristine sulphate. The increase in survival time was thought to arise mainly from 114mIn held by spleen macrophages. The accumulated spleen dose by 7 days after the fifth of the five injections of 74 kBq was 167 Gy. At this dose level the spleen was hypoplastic and the red cells, white cells and platelets in the peripheral blood were severely, though not permanently, suppressed.
Collapse
Affiliation(s)
- L M Cobb
- Division of Experimental Pathology and Therapeutics, Medical Research Council Radiobiology Unit, Chilton, Didcot, Oxon, U.K
| | | |
Collapse
|