1
|
Kılıç G, Demirkan E, Yücel F. Development of Anti-idiotypic Monoclonal Antibody Mimicking SARS-CoV-2 Receptor Binding Domain. Mol Biotechnol 2025; 67:1556-1564. [PMID: 38662257 PMCID: PMC11928402 DOI: 10.1007/s12033-024-01138-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/11/2024] [Indexed: 04/26/2024]
Abstract
Using the hybridoma technique, we developed a panel of anti-idiotypic monoclonal antibodies (aId-mAb) that mimic The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Receptor-Binding Domain (RBD) molecule against Fragment antigen-binding (Fab) of anti-SARS-CoV-2 (S1, RBD) antibodies. Investigated the in vivo and in vitro effects of these aId-mAbs we developed and examined their antigenic mimicry abilities. Among these 12 antibodies, 6 aId-mAbs (designated FY1B4, FY2A6, H9F3, E6G7, FY7E11, and FY8H3) were selected for further characterization in a series of experiments. First, competitive receptor binding assay results confirmed that six aId-mAbs could specifically bind to the ACE2 receptor in target cells and block the interaction between the RBD molecule and the ACE receptor. Moreover, we examined the immunological activities of these aId-mAbs in female BALB/c and showed that E6G7, H7E11, and H8H3 aId-mAbs induce an antibody response by mimicking RBD and stimulating the immune system. It is considered that these three aId-mAbs will be evaluated as SARS-CoV-2 vaccine candidate molecules in future studies.
Collapse
Affiliation(s)
- Gamze Kılıç
- Bursa Uludag University, Faculty of Arts and Sciences, Biology Department, Görükle Campus, Bursa, Turkey
- TUBITAK, Marmara Research Center, Life Sciences, Genetic Engineering and Biotechnology, Kocaeli, Turkey
| | - Elif Demirkan
- Bursa Uludag University, Faculty of Arts and Sciences, Biology Department, Görükle Campus, Bursa, Turkey
| | - Fatıma Yücel
- TUBITAK, Marmara Research Center, Life Sciences, Genetic Engineering and Biotechnology, Kocaeli, Turkey.
| |
Collapse
|
2
|
Anti-GD2 Directed Immunotherapy for High-Risk and Metastatic Neuroblastoma. Biomolecules 2022; 12:biom12030358. [PMID: 35327550 PMCID: PMC8945428 DOI: 10.3390/biom12030358] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroblastoma is one of the few childhood cancers that carries a tumor-specific antigen in the form of a glycolipid antigen known as GD2. It has restricted expression in normal tissue, such as peripheral afferent nerves. Monoclonal antibodies targeting GD2 have been applied clinically to high-risk neuroblastoma with significant success. However, there are different anti-GD2 products and administration regimens. For example, anti-GD2 has been used in combination with chemotherapy during the induction phase or with retinoic acid during the maintenance stage. Regimens also vary in the choice of whether to add cytokines (i.e., IL-2, GMCSF, or both). Furthermore, the addition of an immune enhancer, such as β-glucan, or allogeneic natural killer cells also becomes a confounder in the interpretation. The question concerning which product or method of administration is superior remains to be determined. So far, most studies agree that adding anti-GD2 to the conventional treatment protocol can achieve better short- to intermediate-term event-free and overall survival, but the long-term efficacy remains to be verified. How to improve its efficacy is another challenge. Late relapse and central nervous system metastasis have emerged as new problems. The methods to overcome the mechanisms related to immune evasion or resistance to immunotherapy represent new challenges to be resolved. The newer anti-GD2 strategies, such as bispecific antibody linking of anti-GD2 with activated T cells or chimeric antigen receptor T cells, are currently under clinical trials, and they may become promising alternatives. The use of anti-GD2/GD3 tumor vaccine is a novel and potential approach to minimizing late relapse. How to induce GD2 expression from tumor cells using the epigenetic approach is a hot topic nowadays. We expect that anti-GD2 treatment can serve as a model for the use of monoclonal antibody immunotherapy against cancers in the future.
Collapse
|
3
|
Abstract
The prognosis for advanced neuroblastoma remains poor with high risk of recurrence after consolidation. Therapies based on monoclonal antibodies that specifically target disialoganglioside GD2 on tumor cells are improving treatment results for high-risk neuroblastoma. This article reviews the use of anti-GD2 antibodies either as monotherapy or as part of a larger and more complex treatment approach for advanced neuroblastoma. We review how anti-GD2 antibodies can be combined with other treatments or strategies to enhance their clinical effects. Tumor resistance and other problems that decrease the efficacy of anti-GD2 antibodies are discussed. Future developments in the area of anti-GD2 immunotherapies for neuroblastoma are also addressed.
Collapse
|
4
|
Abstract
Current treatment of malignant melanoma exemplifies not only the need for translational research but also many of the challenges of moving from bench to bedside. Melanoma remains unique among solid tumors in that its treatment primarily is surgical. Radiation is of limited benefit, and chemotherapy has been disappointing in both the adjuvant and metastatic settings. This leaves clinicians with few options for reducing the chance of recurrence after surgery and for treating unresectable disease. With this in mind, there has been a fervent attempt to identify novel approaches to melanoma therapy and translate them into clinical use.
Collapse
|
5
|
Robins RA. Section Review: Oncologic, Endocrine and Metabolic: Antibodies as immunogens for cancer therapy. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.3.12.1233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
6
|
Reinsberg J. Detection of human antibodies generated against therapeutic antibodies used in tumor therapy. Methods Mol Biol 2007; 378:195-204. [PMID: 18605087 DOI: 10.1007/978-1-59745-323-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Application of monoclonal antibodies (MAb) for therapeutic purpose may induce the formation of human antibodies directed against the immunogenic epitopes, which are presented on the therapeutic MAb. Formation of such human antibodies mostly is an undesired side effect, but in the case of newly developed immunotherapeutic tumor treatment strategies it represents the underlying therapeutic effect. Especially the formation of so-called "internal image" antibodies, which are directed against the antigen-combining site (paratope) of the therapeutic antibody, is supposed to evoke specific immune responses against tumor antigens mediated via idiotype-anti-idiotype interactions within the immunoregulatory network. For the monitoring of the immune response after antibody application, the newly formed human antibodies can be measured with immunoassay procedures involving the applied therapeutic antibody as test antibody. Because the original antigen is directed against the therapeutic antibody and inhibits the binding of "internal image" antibodies, a special assay design is needed to avoid interferences with samples containing the antigen. We describe an immunoassay procedure that allows the correct quantification of antiidiotypic antibodies including "internal image" antibodies that are not affected by the original antigen or other serum components that may interact with the therapeutic antibody.
Collapse
Affiliation(s)
- Jochen Reinsberg
- Department of Gynecological Endocrinology and Reproductive Medicine, University of Bonn, Bonn, Germany
| |
Collapse
|
7
|
Wang WWS, Das D, McQuarrie SA, Suresh MR. Design of a bifunctional fusion protein for ovarian cancer drug delivery: single-chain anti-CA125 core-streptavidin fusion protein. Eur J Pharm Biopharm 2006; 65:398-405. [PMID: 17257818 DOI: 10.1016/j.ejpb.2006.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 12/07/2006] [Accepted: 12/08/2006] [Indexed: 10/23/2022]
Abstract
We have developed a universal ovarian cancer cell targeting vehicle that can deliver biotinylated therapeutic drugs. A single-chain antibody variable domain (scFv) that recognizes the CA125 antigen of ovarian cancer cells was fused with a core-streptavidin domain (core-streptavidin-VL-VH and VL-VH-core-streptavidin orientations) using recombinant DNA technology and then expressed in Escherichia coli using the T7 expression system. The bifunctional fusion protein (bfFp) was expressed in a shaker flask culture, extracted from the periplasmic soluble protein, and affinity purified using an IMAC column. The two distinct activities (biotin binding and anti-CA125) of the bfFp were demonstrated using ELISA, Western blot and confocal laser-scanning microscopy (CLSM). The ELISA method utilized human NIH OVCAR-3 cells along with biotinylated bovine serum albumin (B-BSA) or biotinylated liposomes, whereas, the Western blot involved probing with B-BSA. The CLSM study has shown specificity in binding to the OVCAR-3 cell-line. ELISA and Western blot studies have confirmed the bifunctional activity and specificity. In the presence of bfFp, there was enhanced binding of biotinylated antigen and liposome to OVCAR-3 cells. In contrast, the control EMT6 cells, which do not express the CA125 antigen, showed minimal binding of the bfFp. Consequently, bfFp based targeting of biotinylated therapeutic drugs, proteins, liposomes, or nanoparticles could be an alternative, convenient method to deliver effective therapy to ovarian cancer patients. Peritoneal infusion of the bfFp-therapeutic complex could also be effective in locally targeting the most common site of metastatic spread.
Collapse
|
8
|
Pignatari GC, Takeshita D, Parise CB, Soares FA, de Moraes JZ, Han SW. Carcinoembryonic antigen (CEA) mimicry by an anti-idiotypic scFv isolated from anti-Id 6.C4 hybridoma. J Biotechnol 2006; 127:615-25. [PMID: 16989916 DOI: 10.1016/j.jbiotec.2006.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 08/04/2006] [Accepted: 08/15/2006] [Indexed: 11/26/2022]
Abstract
Since carcinoembryonic antigen (CEA) is expressed during embryonic life, it is not immunogenic in humans. The use of anti-idiotypic (Id) antibodies as a surrogate of antigen in the immunization has been considered a promising strategy for breaking tolerance to some tumor associated antigens. We have described an anti-Id monoclonal antibody (MAb), designated 6.C4, which is able to mimic CEA functionally. The anti-Id MAb 6.C4 was shown to elicit antibodies that recognized CEA in vitro and in vivo. In the present study, we sought to verify whether a single chain (scFv) antibody obtained, the scFv 6.C4, would retain the ability to mimic CEA. Two scFv containing the variable heavy and light chain domains of 6.C4 were constructed with a 15-amino acid linker: one with and another without signal peptide. DNA immunization of mice with both forms of scFv individually elicited antibodies able to recognize CEA.
Collapse
|
9
|
Abstract
The need for less toxic adjuvant therapies and a better understanding of the processes by which the immune system can eradicate micrometastatic disease has generated significant interest in breast cancer immunotherapy. There are many potential approaches to stimulating an immune response against a tumor, each with relative advantages and disadvantages in regards to cost, immunogenicity, and clinical applicability in treating breast cancer. This article will review the mechanisms by which the immune system can recognize and eradicate neoplastic cells and the various methods of stimulating an anti-tumor immune response. Obstacles to the clinical effectiveness of immunotherapies in breast cancer are also discussed.
Collapse
Affiliation(s)
- Michael S Sabel
- Department of Surgery, University of Michigan, 3304 Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
10
|
Reinartz S, Köhler S, Schlebusch H, Krista K, Giffels P, Renke K, Huober J, Möbus V, Kreienberg R, DuBois A, Sabbatini P, Wagner U. Vaccination of patients with advanced ovarian carcinoma with the anti-idiotype ACA125: immunological response and survival (phase Ib/II). Clin Cancer Res 2004; 10:1580-7. [PMID: 15014007 DOI: 10.1158/1078-0432.ccr-03-0056] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE A Phase I/IIb multicenter study was conducted to evaluate the safety and immunogenicity of the anti-idiotypic antibody vaccine ACA125 that functionally imitates the tumor antigen CA125 in 119 patients with advanced ovarian carcinoma. A preliminary report on the initial 42 patients demonstrated safety and immunogenicity. EXPERIMENTAL DESIGN Using the complete intention-to-treat population (n = 119) who received a mean of 9.7 ACA125 applications, survival was analyzed with respect to immunological responses. RESULTS In 81 patients (68.1%), a specific anti-anti-idiotypic antibody (Ab3) response could be induced. Additionally, the development of CA125-specific antibodies (Ab1') and antibody-dependent cell-mediated cytotoxicity of CA125-positive tumor cells was observed in 50.4% and 26.9% of patients, respectively. The median survival of all patients was 19.4 months (range, 0.5-56.1 months). Ab3-positive patients showed a significantly longer survival (median, 23.4 months; P < 0.0001) as compared with Ab3-negative patients (median, 4.9 months). A positive Ab3 response remained associated with longer survival when controlling for other prognostic factors including FIGO (International Federation of Gynecologists and Obstetricians) stage, response to and type of first-line chemotherapy, number of previous treatments, or concomitant antitumor therapy. With regard to safety, repeated vaccination was well tolerated. No serious adverse events related to the application of ACA125 occurred. CONCLUSIONS Although the uncontrolled design of this study prevents definitive conclusions with respect to subgroups, the data support a relationship between Ab3 response and survival time. Thus, the need for further randomized, controlled clinical trials to establish efficacy of the vaccine ACA125 seems to be indicated.
Collapse
Affiliation(s)
- Silke Reinartz
- Center of Obstetrics and Gynecology, University of Marburg, Marburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Möbus VJ, Baum RP, Bolle M, Kreienberg R, Noujaim AA, Schultes BC, Nicodemus CF. Immune responses to murine monoclonal antibody-B43.13 correlate with prolonged survival of women with recurrent ovarian cancer. Am J Obstet Gynecol 2003; 189:28-36. [PMID: 12861134 DOI: 10.1067/mob.2003.347] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE We evaluated the therapeutic efficiency of the murine monoclonal antibody-B43.13 in the treatment of patients with recurrent ovarian cancer. STUDY DESIGN This was a retrospective study of immune responses and survival in 44 patients who were treated with technetium 99m-labeled monoclonal antibody-B43.13, a murine monoclonal antibody that is directed against the tumor-associated antigen CA125. Most patients were pretreated heavily. Biologic activity was quantified by the assay of immune responses to the human anti-murine antibodies against the monoclonal antibody-B43.13 variable region (Ab(2)) and antibodies that target the CA 125 antigen itself (anti-CA 125 antibody). RESULTS More than one half of patients (56.8%) survived for >12 months after the first dose of monoclonal antibody B43.13; 34.1% of the patients survived >24 months. To date, 6 of the 44 patients are alive, with survival times of 4 to 7.5 years after the start of the antibody treatment. More than 60% of the evaluable patients met predefined criteria for robust, treatment-emergent human anti-murine antibodies and Ab(2) responses, and these responses were associated with improved survival rates. Median survival time increased approximately 3-fold for human anti-murine antibody responders (22.6 months) versus nonresponders (7.2 months; P <.0016, log-rank test) and 2-fold for Ab(2) responders (18.3 months) versus nonresponders (9.3 months). No serious drug-associated adverse events were reported. CONCLUSION The associations between multiple types of immune response and improved clinical outcomes suggest that monoclonal antibody-B43.13 should be further evaluated for potential use as an immunotherapy for CA125-expressing malignancies.
Collapse
Affiliation(s)
- Volker J Möbus
- Department of Obstetrics and Gynecology, Städtische Kliniken, Frankfurt, Germany.
| | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Patients presenting with thick primary melanomas or those with regional nodal metastases have a high risk of recurrence after surgery alone. Chemotherapy has limited efficacy in the adjuvant setting, and while the use of high-dose interferon in the adjuvant setting has been reported to improve survival, treatment with interferon is not without significant cost and toxicity. Mounting evidence suggests a prominent role for the immune system in the natural history of melanoma, and the clinical success of interferon highlights the potential for immunotherapy to prevent recurrence. Many researchers hope to use melanoma vaccines to reduce recurrence without significant toxicity, and many different vaccine strategies are under investigation. Peptide vaccines attempt to induce immunity to melanoma MHC-restricted peptide antigens by delivering the peptide to the patient along with an immune adjuvant meant to induce inflammation and stimulate immunity. While peptide vaccines have advantages with regard to cost and feasibility, it is still unclear whether highly purified peptides will stimulate an adequate immune response. An alternative approach is the use of cellular vaccines. Autologous cellular vaccines present all biologically relevant antigens to the immune system, but this is limited to individuals with sufficient tumor to prepare a vaccine. Allogeneic cellular vaccines are based on the fact that melanoma-associated antigens are shared among a large number of patients, so a vaccine prepared from a cultured cell line could stimulate an anti-tumor immune response in many patients. Allogeneic vaccines are available for all patients, and can be standardized, preserved and distributed in a manner akin to any other therapeutic agent. Because of this, they are more readily available for evaluation in large trials, and there are two major allogeneic vaccines presently being evaluated as an adjuvant therapy for melanoma. Several additional approaches to vaccine therapies are being investigated including among others ganglioside vaccines, viral oncolysates, cytokine gene-modified tumor cell vaccines, dendritic cell vaccines, anti-idiotype antibodies and DNA vaccines. While there appears to be tremendous potential for vaccines, it must be remembered that there has been significant interest in immunotherapy for melanoma for over 50 years and, to date, no large prospective, randomized trial has shown a survival benefit.
Collapse
Affiliation(s)
- Michael S Sabel
- University of Michigan Comprehensive Cancer Center, Ann Arbor 48109, USA.
| | | |
Collapse
|
13
|
Chang X, Cui H, Feng J, Li Y, Liu B, Cao S, Cheng Y, Qian H. Preparation of humanized ovarian carcinoma anti-idiotypic minibody. HYBRIDOMA AND HYBRIDOMICS 2003; 22:109-15. [PMID: 12831536 DOI: 10.1089/153685903321948030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Murine anti-idiotypic monoclonal antibody (MAb) 6B11 mimicking the tumor-associated antigen OC166-9 is used as a vaccine for the induction of an anti-tumoral immunity in experiments of in vitro and in vivo animal model with ovarian carcinoma. In this article, we have humanized 6B11 anti-idiotypic minibody using overlap polymerase chain reaction (PCR) and DNA recombinant technique, prokaryotic expression vector was produced by genetic fusion of 6B11V(L)-V(H) to human IgG1 hinge and CH3 region. Transformed E. coli BL21(DE3) were propagated and induced by isopropyl-beta D-thiogalactopyranoside (IPTG). Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) showed that a protein band with molecular weight of 50kD appeared as the expected size after transformation. Molecular weight of 100 kDa may be examined by electrophoresis in nondenaturing systems. The fusion protein was analyzed with enzyme-linked immunosorbant assay (ELISA), inhibition ELISA tests and Western blot, respectively. The humanized anti-idiotype minibody showed capacity of bivalent binding to ovarian cancer MAb COC166-9 and goat anti-human immunoglobulin IgG1. It is useful reagents for clinical use.
Collapse
Affiliation(s)
- Xiaohong Chang
- Gynecologic Oncology Center, People's Hospital, Peking University, Beijing 100044, China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Pawlik TM, Sondak VK. Malignant melanoma: current state of primary and adjuvant treatment. Crit Rev Oncol Hematol 2003; 45:245-64. [PMID: 12633838 DOI: 10.1016/s1040-8428(02)00080-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Metastatic malignant melanoma remains a highly lethal disease with an incidence that continues to rise. Management of melanoma includes definitive local, regional and distant control. There is substantial prospective and retrospective data to base the extent of both primary as well as adjuvant therapy. The results of these trials have on occasion been at odds. A critical assessment of the available information pertaining to the adjuvant treatment of cutaneous melanoma is needed. This review provides a critical assessment of the current data that is available to guide both primary resection as well as adjuvant therapy. To date, current trials have shown little promise with nonspecific immunostimulants and cytotoxic chemotherapy. In contrast, dose interferon-alpha2b has been shown to improve relapse-free survival and likely improves melanoma-specific survival as well. Based on the available data, interferon-alpha2b remains the adjuvant therapy of choice for high-risk patients treated outside clinical trials, and the appropriate control arm for clinical trials evaluating new or modified adjuvant regimens.
Collapse
Affiliation(s)
- Timothy M Pawlik
- Division of Surgical Oncology, University of Michigan Medical School, Ann Arbor, MI 48109-0031, USA
| | | |
Collapse
|
15
|
Reinartz S, Boerner H, Koehler S, Von Ruecker A, Schlebusch H, Wagner U. Evaluation of immunological responses in patients with ovarian cancer treated with the anti-idiotype vaccine ACA125 by determination of intracellular cytokines--a preliminary report. Hybridoma (Larchmt) 1999; 18:41-5. [PMID: 10211787 DOI: 10.1089/hyb.1999.18.41] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In a first clinical trial, 45 patients with advanced ovarian carcinoma and recurrences were treated with the murine monoclonal anti-idiotypic antibody (Ab2) designated ACA125 for active immunotherapy. The monoclonal antibody (MAb) ACA125 mimics a specific epitope of the tumor-associated antigen CA125 expressed by most malignant ovarian tumors. Patients with CA125-positive tumors are immunologically tolerant to CA125, which could be overcome by the use of an anti-idiotypic antibody as a surrogate for the tumor antigen CA125. An immunological response to the anti-idiotype ACA125 in these patients was associated with a statistically significant survival prolongation. Humoral immunity to ACA125 was assessed by induction of anti-anti-idiotypic antibodies (Ab3) directed against CA125. Using flow cytometric detection methods we observe alterations of the intracellular cytokines IFN-gamma, IL-2, and IL-4 at the single-cell level during the course of immunization. There was a strong increase of intracellular IFN-gamma and IL-2 characteristic for a Th1 cell type immune response after treatment with ACA125. A delayed induction of Th2 type response, which promotes antibody-mediated immunity by B cells, could also be detected. The understanding of the kinetics of Th1 and Th2 responses could be important to improve treatment schedules for effective immunotherapy with anti-idiotype vaccines.
Collapse
Affiliation(s)
- S Reinartz
- Center of Obstetrics and Gynecology, University of Bonn, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Schultes BC, Zhang C, Xue LY, Noujaim AA, Madiyalakan R. Immunotherapy of human ovarian carcinoma with OvaRex MAb-B43.13 in a human-PBL-SCID/BG mouse model. Hybridoma (Larchmt) 1999; 18:47-55. [PMID: 10211788 DOI: 10.1089/hyb.1999.18.47] [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: 11/12/2022]
Abstract
The monoclonal antibody (MAb) B43.13, binding to the ovarian cancer-associated antigen CA125, has been injected into more than 200 patients with ovarian cancer to detect recurrence of the disease. The follow-up of the patients revealed surprisingly long survival spans for several patients despite high CA125 levels. To investigate the therapeutic effectiveness of OvaRex MAb-B43.13 (AltaRex, Edmonton, Canada) under well-controlled conditions, the antibody was tested in a human-PBL-SCID/BG mouse model with CA125 positive human ovarian cancer cells. Mice were reconstituted with human peripheral blood lymphocytes (PBL, normal donors) by intraperitoneal (IP) injection of 2 to 3 x 10(7) PBL/mouse. OvaRex MAb-B43.13 was administered at 100 microg/mouse in phosphate buffered saline (PBS), in three different experimental set-ups. An isotype-matched control antibody (MOPC21 or MAb-170) and PBS injection served as controls. The ovarian cancer cell line NIH:OVCAR-NU-3 was injected IP at 1 x 10(6) cells/mouse or subcutaneously (SC) at 4 x 10(6) cells/mouse. Human-PBL-SCID/BG mice were either immunized before injection of tumor cells, along with tumor cells or after small tumors were established (2 weeks after transplantation). Antibody injections were repeated twice in 2-week intervals. Functional and cellular characterization of serum and PBL from these mice demonstrated the successful engraftment of a human immune system in those mice. All three experiments showed that OvaRex MAb-B43.13 treatment could (a) delay or prevent development of tumors; (b) reduce the size of small established tumors (SC tumor injection) or suppress ascites formation; (c) delay tumor growth when injected prior to tumor implantation; and (d) prolong the survival of the mice (i.p. tumor injection).
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/therapeutic use
- CA-125 Antigen/immunology
- Disease Models, Animal
- Female
- Flow Cytometry
- Humans
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Immunohistochemistry
- Immunotherapy
- Mice
- Mice, SCID
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/therapy
- Tumor Cells, Cultured
Collapse
|
17
|
Lou D, Kohler H. Enhanced molecular mimicry of CEA using photoaffinity crosslinked C3d peptide. Nat Biotechnol 1998; 16:458-62. [PMID: 9592395 DOI: 10.1038/nbt0598-458] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antigen mimicry of using anti-idiotypic antibodies for use as cancer vaccines has been disappointing due to the weak immunogenicity of immunoglobulin variable domains. To enhance the immunogenicity of an anti-idiotype vaccine we incorporated a molecular adjuvant peptide into the antibody. The peptide is derived from the C3d region known to bind CR2 receptors on B-cells. A photoreactive peptide is synthesized that affinity-labels a single site in the antibody variable domain. The molecular adjuvant peptide is crosslinked to the anti-idiotype mimetic by chemical means without modifying other sites on the antibody. The C3d-conjugated anti-idiotype antibody induces a strong idiotype and antigen-specific response in mice.
Collapse
Affiliation(s)
- D Lou
- Immpheron, Inc., Lexington, KY 40509, USA
| | | |
Collapse
|
18
|
|
19
|
Reinsberg J, Krebs D. Are human anti-idiotypic anti-OC125 antibodies formed after immunization with the anti-CA125 antibody B43.13? Hybridoma (Larchmt) 1997; 16:59-63. [PMID: 9085130 DOI: 10.1089/hyb.1997.16.59] [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: 02/04/2023]
Abstract
The two monoclonal antibodies (MAb) OC125 and B43.13 both recognize antigenic determinants on the cancer antigen 125 (CA125) molecule. The aim of the present study was to clarify how far antibodies formed by patients treated with B43.13 may cross-react with idiotopes of OC125. Serum samples from 15 ovarian cancer patients treated with B43.13 were tested for the presence of anti-idiotypic anti-OC125 antibodies; 4 of these patients previously had received OC125 F(ab')2 fragments. Six patients treated only with B43.13 and all patients pretreated with OC125 fragments developed a considerable increase of human anti-mouse anti-bodies after B43.13 infusion, indicating an immune response to antibody infusion. However, none of the patients treated only with B43.13 developed detectable levels of anti-idiotypic anti-OC125 antibodies, whereas in 2 patients pretreated with OC125 fragments, the pre-existing concentration of anti-idiotypic anti-OC125 antibodies further increased after B43.13 infusion. The binding of these newly formed antibodies to OC125 was not inhibited by the B43.13, but about 70% of the binding was inhibited by the CA125 antigen. The present data suggest that the idiotopes expressed on the antibodies B43.13 and OC125, respectively, are completely different. Thus, anti-idiotypic anti-B43.13 antibodies may not cross-react with OC125. A greater number of cases is needed to clarify how far the increase of anti-idiotypic anti-OC125 antibodies observed in 2 patients pretreated with OC125 fragments really is due to B43.13 infusion.
Collapse
Affiliation(s)
- J Reinsberg
- Department of Gynecology and Obstetrics, University of Bonn, Germany
| | | |
Collapse
|
20
|
Holz E, Raab R, Riethmüller G. Antibody-based immunotherapeutic strategies in colorectal cancer. Recent Results Cancer Res 1996; 142:381-400. [PMID: 8893351 DOI: 10.1007/978-3-642-80035-1_21] [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: 05/22/2023]
Abstract
Monoclonal antibodies may well be on their way to becoming an integral part of therapy after the most recent success in prolonging overall and recurrence-free survival in patients with stage III colorectal cancer after potentially curative surgery. After a median follow-up of 5 years, antibody treatment reduced the overall death rate by 30% and decreased the recurrence rate by 27%. These results are similar with regard to efficacy but there is less toxicity with those obtained in contemporary and more recent chemotherapy trials. The key to success with high-molecular-weight substances such as immunoglobulines lies in the careful selection of the appropriate target population, i.e., patients with minimal residual disease, where only isolated tumor cells which are readily accessible to therapy are present. An argument for combining immunotherapy with chemotherapy can be made on the basis of the phenotype of individual disseminated tumor cells, which by immunocytochemistry were found to only rarely express proliferation-associated antigens and therefore are independent of the cell cycle. Further efforts to improve immunotherapy have also led to the combined clinical use of antibodies with biologic response modifiers which are known to enhance effector cell-mediated antibody-dependent cytotoxicity. Additional rationally designed clinical trials are ongoing in which specific immunotherapy is directed towards known, readily accessible, and abundant cell target structures, either alone or combined with treatment modalities which employ different action mechanisms.
Collapse
Affiliation(s)
- E Holz
- Tumorzentrum München, Medizinischen Fakultäten der Ludwig-Maximilians-Universität, Technischen Universität, Germany
| | | | | |
Collapse
|
21
|
Hodge JW, Schlom J, Donohue SJ, Tomaszewski JE, Wheeler CW, Levine BS, Gritz L, Panicali D, Kantor JA. A recombinant vaccinia virus expressing human prostate-specific antigen (PSA): safety and immunogenicity in a non-human primate. Int J Cancer 1995; 63:231-7. [PMID: 7591210 DOI: 10.1002/ijc.2910630215] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Prostate-specific antigen (PSA) is a serine protease secreted by prostatic epithelial cells and is widely used as a marker for prostate cancer. The tissue specificity of PSA makes it a potential target for active specific immunotherapy, especially in prostate cancer patients who have undergone prostatectomy and in whom the only PSA-expressing tissue in the body resides in metastatic deposits. We report here the cloning, construction and immunological consequences of immunization of rhesus monkeys with a recombinant vaccinia virus expressing human PSA (designated rV-PSA). The prostate gland of the rhesus is structurally and functionally similar to the human prostate. While rodent and other mammalian species do not share homology with human PSA, there is 94% homology between the amino acid sequences of rhesus and human PSA. Immunization of rhesus monkeys with wild-type vaccinia virus or rV-PSA elicited the usual low-grade constitutional symptoms of vaccinia virus infection. There was no evidence of any adverse effects in any immunized monkeys. A short-lived PSA-specific IgM antibody response was noted in all rV-PSA immunized monkeys regardless of dose level. All monkeys receiving the 10(8)pfu dose of rV-PSA demonstrated PSA-specific T-cell responses that were maintained up to 270 days. No differences in anti-PSA immune responses or toxicity were observed in animals that received prostatectomy prior to immunization. Our results thus demonstrate the safety and immunogenicity of rV-PSA in a non-human primate and have implications for potential specific immunotherapy protocols using PSA as a target.
Collapse
Affiliation(s)
- J W Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Madiyalakan R, Sykes TR, Dharampaul S, Sykes CJ, Baum RP, Hör G, Noujaim AA. Antiidiotype induction therapy: evidence for the induction of immune response through the idiotype network in patients with ovarian cancer after administration of anti-CA125 murine monoclonal antibody B43.13. Hybridoma (Larchmt) 1995; 14:199-203. [PMID: 7590780 DOI: 10.1089/hyb.1995.14.199] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The immune status of ovarian cancer patients receiving anti-CA125 murine monoclonal antibody B43.13 was evaluated by measuring antiidiotypic antibodies (Ab2), antiantiidiotypic antibodies (Ab3), antiisotypic human antimouse antibodies (HAMA), interferon-gamma, and CA125 levels in the serum. A specific assay was developed for the determination of Ab2 antibodies using chimeric MAb B43.13. Of the 50 patients studied, 26 had elevated levels of Ab2. Eleven of these 26 patients also had high titer of antiantiidiotypic (Ab3) antibodies. Eight of the 22 patients analyzed had increased interferon-gamma levels. A tentative correlation was found between survival of these patients' antiidiotype induction.
Collapse
Affiliation(s)
- R Madiyalakan
- Biomira Research Inc., University of Alberta, Edmonton, Canada
| | | | | | | | | | | | | |
Collapse
|
24
|
Hebbar M, Vanlemmens L, Lecomte S, Bonneterre J. [Immunotherapy of malignant melanoma. New prospects]. Rev Med Interne 1995; 16:260-70. [PMID: 7746965 DOI: 10.1016/0248-8663(96)80705-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Malignant melanoma is an immunogenic tumor which can induce host humoral and cellular responses, and is a good model for development of anti-cancer immunotherapy. During the last decade systemically-administrated interferon and interleukin-2 have been used. Advances in immunology and molecular biology could allow a more specific and active immunotherapy. Perspectives include chemo-immunotherapy, monoclonal antibodies alone or in combination with cytotoxic agents, adoptive immunotherapy with tumor-infiltrating lymphocytes (TIL), gene therapy designed to increase tumor immunogenicity, and active immunotherapy with vaccines.
Collapse
Affiliation(s)
- M Hebbar
- Service de l'hôpital de jour, centre Oscar-Lambret, Lille, France
| | | | | | | |
Collapse
|