1
|
Hein V, Baeza-Kallee N, Bertucci A, Colin C, Tchoghandjian A, Figarella-Branger D, Tabouret E. GD3 ganglioside is a promising therapeutic target for glioma patients. Neurooncol Adv 2024; 6:vdae038. [PMID: 38590763 PMCID: PMC11000324 DOI: 10.1093/noajnl/vdae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Glioblastoma is the most frequent and aggressive primary brain tumor in adults. Currently, no curative treatment is available. Despite first-line treatment composed by the association of surgery, radiotherapy, and chemotherapy, relapse remains inevitable in a median delay of 6 to 10 months. Improving patient management and developing new therapeutic strategies are therefore a critical medical need in neuro-oncology. Gangliosides are sialic acid-containing glycosphingolipids, the most abundant in the nervous system, representing attractive therapeutic targets. The ganglioside GD3 is highly expressed in neuroectoderm-derived tumors such as melanoma and neuroblastoma, but also in gliomas. Moreover, interesting results, including our own, have reported the involvement of GD3 in the stemness of glioblastoma cells. In this review, we will first describe the characteristics of the ganglioside GD3 and its enzyme, the GD3 synthase (GD3S), including their biosynthesis and metabolism. Then, we will detail their expression and role in gliomas. Finally, we will summarize the current knowledge regarding the therapeutic development opportunities against GD3 and GD3S.
Collapse
Affiliation(s)
- Victoria Hein
- Aix-Marseille Université, CNRS, INP, Inst Neurophysiopathol, GlioME Team, Marseille, France
| | - Nathalie Baeza-Kallee
- Aix-Marseille Université, CNRS, INP, Inst Neurophysiopathol, GlioME Team, Marseille, France
- Aix-Marseille Univ, Réseau Préclinique et Translationnel de Recherche en Neuro-oncologie PETRA, Plateforme PETRA“TECH” and Plateforme PE”TRANSLA,”Marseille, France
| | - Alexandre Bertucci
- Aix-Marseille Université, CNRS, INP, Inst Neurophysiopathol, GlioME Team, Marseille, France
- APHM, CHU Timone, Service de Neuro-Oncologie, MarseilleFrance
| | - Carole Colin
- Aix-Marseille Université, CNRS, INP, Inst Neurophysiopathol, GlioME Team, Marseille, France
- Aix-Marseille Univ, Réseau Préclinique et Translationnel de Recherche en Neuro-oncologie PETRA, Plateforme PETRA“TECH” and Plateforme PE”TRANSLA,”Marseille, France
| | - Aurélie Tchoghandjian
- Aix-Marseille Université, CNRS, INP, Inst Neurophysiopathol, GlioME Team, Marseille, France
- Aix-Marseille Univ, Réseau Préclinique et Translationnel de Recherche en Neuro-oncologie PETRA, Plateforme PETRA“TECH” and Plateforme PE”TRANSLA,”Marseille, France
| | | | - Emeline Tabouret
- Aix-Marseille Université, CNRS, INP, Inst Neurophysiopathol, GlioME Team, Marseille, France
- APHM, CHU Timone, Service de Neuro-Oncologie, MarseilleFrance
- Aix-Marseille Univ, Réseau Préclinique et Translationnel de Recherche en Neuro-oncologie PETRA, Plateforme PETRA“TECH” and Plateforme PE”TRANSLA,”Marseille, France
| |
Collapse
|
2
|
Tong W, Maira M, Roychoudhury R, Galan A, Brahimi F, Gilbert M, Cunningham AM, Josephy S, Pirvulescu I, Moffett S, Saragovi HU. Vaccination with Tumor-Ganglioside Glycomimetics Activates a Selective Immunity that Affords Cancer Therapy. Cell Chem Biol 2019; 26:1013-1026.e4. [DOI: 10.1016/j.chembiol.2019.03.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/19/2018] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
|
3
|
Casey DL, Lin TY, Cheung NKV. Exploiting Signaling Pathways and Immune Targets Beyond the Standard of Care for Ewing Sarcoma. Front Oncol 2019; 9:537. [PMID: 31275859 PMCID: PMC6593481 DOI: 10.3389/fonc.2019.00537] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Ewing sarcoma (ES) family of tumors includes bone and soft tissue tumors that are often characterized by a specific translocation between chromosome 11 and 22, resulting in the EWS-FLI1 fusion gene. With the advent of multi-modality treatment including cytotoxic chemotherapy, surgery, and radiation therapy, the prognosis for patients with ES has substantially improved. However, a therapeutic plateau is now reached for both localized and metastatic disease over the last two decades. Burdened by the toxicity limits associated with the current frontline systemic therapy, there is an urgent need for novel targeted therapeutic strategies. In this review, we discuss the current treatment paradigm of ES, and explore preclinical evidence and emerging treatments directed at tumor signaling pathways and immune targets.
Collapse
Affiliation(s)
- Dana L Casey
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Tsung-Yi Lin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
4
|
Dobrenkov K, Ostrovnaya I, Gu J, Cheung IY, Cheung NKV. Oncotargets GD2 and GD3 are highly expressed in sarcomas of children, adolescents, and young adults. Pediatr Blood Cancer 2016; 63:1780-5. [PMID: 27304202 PMCID: PMC5215083 DOI: 10.1002/pbc.26097] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND GD2 and GD3 are the tumor-associated glycolipid antigens found in a broad spectrum of human cancers. GD2-specific antibody is currently a standard of care for high-risk neuroblastoma therapy. In this study, the pattern of GD2 and GD3 expression among pediatric/adolescent or young adult tumors was determined, providing companion diagnostics for targeted therapy. METHODS Ninety-two specimens of human osteosarcoma (OS), rhabdomyosarcoma (RMS), Ewing family of tumors, desmoplastic small round cell tumor (DSRCT), and melanoma were analyzed for GD2/GD3 expression by immunohistochemistry. Murine monoclonal antibody 3F8 was used for GD2 staining, and R24 for GD3. Staining was scored according to both intensity and percentage of positive tumor cells from 0 to 4. RESULTS Both gangliosides were highly prevalent in OS and melanoma. Among other tumors, GD3 expression was higher than GD2 expression. Most OS samples demonstrated strong staining for GD2 and GD3, whereas expression for other tumors was highly variable. Mean intensity of GD2 expression was significantly more heterogeneous (P < 0.001) when compared to GD3 across tumor types. When assessing the difference between GD2 and GD3 expression in all tumor types combined, GD3 expression had a significantly higher score (P = 0.049). When analyzed within each cancer, GD3 expression was significantly higher only in DSRCT (P = 0.002). There was no statistical difference in either GD2 or GD3 expression between primary and recurrent sarcomas. CONCLUSION GD2/GD3 expression among pediatric solid tumors is common, albeit with variable level of expression. Especially for patients with sarcoma, these gangliosides can be potential targets for antibody-based therapies.
Collapse
Affiliation(s)
| | - Irina Ostrovnaya
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jessie Gu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Irene Y. Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
,Correspondence to: Nai-Kong V. Cheung, MD, PhD, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, Tel.: 646-888-2313, Fax: 631-422-0452,
| |
Collapse
|
5
|
Rabu C, McIntosh R, Jurasova Z, Durrant L. Glycans as targets for therapeutic antitumor antibodies. Future Oncol 2012; 8:943-60. [PMID: 22894669 DOI: 10.2217/fon.12.88] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glycans represent a vast class of molecules that modify either proteins or lipids. They exert and regulate important and complex functions in both normal and cancer cell metabolism. As such, the most immunogenic glycans have been targeted in passive and active immunotherapy in human cancer for the past 25 years but it is only recently that techniques have become available to uncover novel glycan targets. The main focus of this review article is to highlight why and how monoclonal antibodies (mAbs) recognizing glycans, and in particular the glycans expressed on glycolipids, are being used in various strategies to target and kill cancer cells. The article reports on the historical use of mAbs and on very recent progress made in antitumor therapy using the anti-GD2 mAb and the antiganglioside mAbs, anti-N-glycolylneuraminic acid mAb and anti-Lewis mAb. Anti-GD2 is showing great promise in Phase III clinical trials in adjuvant treatment of neuroblastoma. Racotumomab, an anti-idiotypic mAb mimicking N-glycolylneuraminic acid-containing gangliosides, is currently being tested in a randomized, controlled Phase II/III clinical trial. This article also presents various strategies used by different groups to develop mAbs against these naturally poorly immunogenic glycans.
Collapse
Affiliation(s)
- Catherine Rabu
- Academic Department of Clinical Oncology, City Hospital Campus, University of Nottingham, Nottingham, NG5 1PB, UK
| | | | | | | |
Collapse
|
6
|
Demierre MF, Sabel MS, Margolin KA, Daud AI, Sondak VK. State of the science 60th anniversary review: 60 Years of advances in cutaneous melanoma epidemiology, diagnosis, and treatment, as reported in the journal Cancer. Cancer 2008; 113:1728-43. [PMID: 18798543 DOI: 10.1002/cncr.23643] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Marie-France Demierre
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
7
|
Milner RJ, Salute M, Crawford C, Abbot JR, Farese J. The immune response to disialoganglioside GD3 vaccination in normal dogs: a melanoma surface antigen vaccine. Vet Immunol Immunopathol 2006; 114:273-84. [PMID: 17027091 DOI: 10.1016/j.vetimm.2006.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Revised: 08/14/2006] [Accepted: 08/23/2006] [Indexed: 11/19/2022]
Abstract
As a result of its metastatic potential, canine malignant melanoma like its human counterpart like its human counter part, has a poor response to conventional treatment protocols. This prompted us to investigate the possibility of enhancing the immune response against the melanoma cell surface antigen, disialoganglioside GD3. Initially a flow cytometric study was designed in which the incidence of GD3 on the cell surface, recognized by the monoclonal antibody Mel-1 (R24), was established in canine melanoma cell lines. Results from the flow cytometry found GD3 to be highly expressed (94.2%) in six out of seven canine melanoma cell lines. Since it was thus potentially a good target, a study in which normal dogs were vaccinated intradermally with a vaccine containing GD3 plus adjuvants was designed. The adjuvant included CpG oligodeoxynucleotide (CpG-ODN) sequences and RIBI-adjuvant, which are known to target toll-like receptors (TLR) of the innate immune system. From a cohort of 10 dogs, 4 were vaccinated 3 times, at 4 weekly intervals with GD3 plus adjuvant, and 4 received only RIBI-adjuvant, and 2 phosphate buffered saline. Caliper measurements were collected to assess skin reaction at the vaccination site and sera assayed for IgM and IgG antibodies against GD3 and cell-mediated cytotoxicity against a melanoma cell line. Results from the study found significant differences (P<0.05) in the vaccine site reactions, IgM/IgG levels and cell-mediated cytotoxicity in the vaccinated versus unvaccinated dogs. The addition of CpG-ODN sequences and increasing GD3 concentration in the vaccine increased the inflammation response at the injection site. GD3 IgG and IgM antibodies in vaccinated dogs showed increasing titers over time and achieved significance at weeks 9 and 12, respectively. Cell-mediated cytotoxicity was only detected in peripheral blood mononuclear cells from vaccinated dogs. In conclusion, by combining the tumor antigen GD3 (a known weak self-antigen) and an adjuvant, tolerance was overcome by an innate and adaptive immune response in this population of normal dogs.
Collapse
Affiliation(s)
- R J Milner
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Florida, USA.
| | | | | | | | | |
Collapse
|
8
|
Zou W, Borrelli S, Gilbert M, Liu T, Pon RA, Jennings HJ. Bioengineering of Surface GD3 Ganglioside for Immunotargeting Human Melanoma Cells. J Biol Chem 2004; 279:25390-9. [PMID: 15047693 DOI: 10.1074/jbc.m402787200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
N-Propionyl, N-butyryl (N-Bu), and N-benzoyl mannosamine, as precursors of sialic acid biosynthesis, were incubated with human melanoma SK-MEL-28 cells and resulted in the replacement of N-acetyl groups on the cell surface sialic acid residues, including those associated with GD3. Meanwhile, vaccines containing GD3 and modified GD3 tetrasaccharide-keyhole limpet hemocyanin conjugates were synthesized, and BALB/c mice were immunized with them together with monophosphoryl lipid A adjuvant. The GD3Bu-keyhole limpet hemocyanin conjugate raised the highest IgG titers without any cross-reactivity to unmodified GD3. Expression of GD3Bu epitopes on the surface of SK-MEL-28 cells was confirmed in vitro and in vivo by the binding of a polyclonal antiserum and monoclonal antibody (mAb) 2A, both of which specifically recognize GD3Bu, and by mass spectroscopic analysis of glycolipids extracted from cells. Following expression of GD3Bu on the surface of SK-MEL-28 cells, the cells could be lysed by mAb 2A and GD3Bu antiserum in the presence of complement. Although less effective in the control of existing large size tumors ( approximately 10 mm inner diameter) on BALB/c nu/nu mice, mAb 2A in combination with ManNBu effectively protected mice from SK-MEL-28 tumor grafting. This approach may provide a method to augment the immunogenicity of sialylated human antigens and to avoid generating an autoimmune response to them at same time.
Collapse
Affiliation(s)
- Wei Zou
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario K1A 0R6, Canada
| | | | | | | | | | | |
Collapse
|
9
|
Abstract
Gangliosides, sialic acid-containing glycosphingolipids, have engendered great interest for more than 20 years in the search for target molecules of relevance for tumour growth and formation of metastases and as potential targets for immunotherapy. These molecules show large quantitative and structural variability, which is related to cell type and developmental stage. Their potential role in the formation of tumour metastases was suggested from data supporting that they are involved in cell growth regulation and in cell-cell and cell-matrix adhesion. Moreover, gangliosides are expressed on the cell surface and thereby are accessible for antibodies or other ganglioside-binding molecules to induce cell death, inhibit cell growth and/or inhibit formation of tumour metastasis. All tumours exhibit aberrant ganglioside expression. This includes overexpression of normal ganglioside constituents, which appears to be common among various tumours, and expression of gangliosides not found in normal adult tissue but often found during fetal development. The ganglioside composition of melanoma cells has been found to correlate with their metastatic potential and also to be selectively expressed in cells of a tumour mass and invading tumour cells. Passive immunotherapy using murine or murine/human chimeric monoclonal antiganglioside antibodies in their native form or combined with various effector molecules has been investigated. However, the vaccination strategy using native or structurally modified tumour-associated gangliosides in combination with adjuvants is currently the dominant method in clinical trials. The outcomes reported so far vary between type of tumour and treatment strategies. However, we believe that targeting gangliosides is as promising as any other immune therapeutic strategy, and basic research as well as clinical trials utilising new aspects is encouraged.
Collapse
Affiliation(s)
- Pam Fredman
- Experimental Neuroscience Section, Institute of Clinical Neuroscience, The Sahlgrenska Institute at Göteborg University, Sahlgrenska University Hospital, SE 43180 Molndal, Sweden.
| | | | | |
Collapse
|
10
|
Abrams K, Yunusov MY, Slichter S, Moore P, Nelp WB, Burstein SA, McDonough S, Durack L, Storer B, Storb R, Gass MJ, Georges G, Nash RA. Recombinant human macrophage colony-stimulating factor-induced thrombocytopenia in dogs. Br J Haematol 2003; 121:614-22. [PMID: 12752103 DOI: 10.1046/j.1365-2141.2003.04313.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To characterize recombinant human macrophage-colony stimulating factor (rhM-CSF)-associated thrombocytopenia (TCP), in vivo studies were performed in dogs, including the biodistributions and recoveries of radiolabelled autologous and allogeneic platelets. rhM-CSF induced a reversible, dose-dependent decrease in platelet counts. The number of megakaryocytes in spleen and marrow of rhM-CSF-treated dogs was increased two to threefold. Recoveries of allogeneic platelets transfused from rhM-CSF-treated donors into tolerized recipients (n = 3) were not significantly different from allogeneic baseline studies (93 +/- 10% of baseline values at 24 h and 90 +/- 1% at 40 h), whereas autologous platelets infused back into rhM-CSF-treated donors had decreased recoveries (45 +/- 2% of baseline values at 24 h, P = 0.03 and 20 +/- 4% at 40 h, P = 0.001). Platelet biodistribution studies showed increased accumulation of radiolabelled platelets over the spleens and livers of rhM-CSF-treated dogs. Histochemistry showed increased levels of platelet-specific antigen (CD41; glycoprotein IIb) associated with Kupffer cells. The sensitivity of platelets from rhM-CSF-treated dogs to activation from thrombin, as measured by expression of P-selectin (CD62P), was not significantly different when compared with baseline studies (P = 0.18; n = 4). These results support the concept that rhM-CSF induces an activation of the monocyte-macrophage system (MMS), which causes a reversible TCP in a dog model.
Collapse
Affiliation(s)
- Kraig Abrams
- Puget Sound Blood Center, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109-1024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
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: 1.6] [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
|
12
|
|
13
|
Cunto-Amesty G, Luo P, Monzavi-Karbassi B, Kieber-Emmons T. Exploiting molecular mimicry: defining rules of the game. Int Rev Immunol 2002; 20:157-80. [PMID: 11878763 DOI: 10.3109/08830180109043032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Molecular mimicry has been touted as a mean to develop new generation of vaccines to target carbohydrate antigens on pathogens and on tumor cells. Structural and immunological rules governing molecular mimicry require definition for its successful exploitation. Of interest are the kinds of structures that peptides adopt as carbohydrate mimics, the extent to which topological or sequence similarities among peptide mimeotopes define serum cross-reactivity to carbohydrate antigens and the extent to which peptide mimeotopes affect T-cell responses. Rational design concepts can be applied to define how a peptide may mimic carbohydrate antigens, similarities in binding affinities of antibodies for carbohydrate and for peptides, how peptides can mimic core structures on otherwise dissimilar carbohydrate antigens, and how peptide mimeotopes can be used to manipulate cellular responses not achievable by carbohydrate antigens.
Collapse
Affiliation(s)
- G Cunto-Amesty
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | |
Collapse
|
14
|
Koh Y, Tsunoda T, Iwahashi M, Yamaue H, Ishimoto K, Tanimura H, Fukumoto H, Nakamura T, Tatsumi Y, Shimizu M, Saijo N, Nishio K. Decreased expression of alpha2,8 sialyltransferase and increased expression of beta1,4 N-acetylgalactosaminyltransferase in gastrointestinal cancers. Exp Biol Med (Maywood) 2002; 227:196-200. [PMID: 11856818 DOI: 10.1177/153537020222700307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Gangliosides such as GD3, GM2, and GD2 are abundantly expressed on the cell surfaces of various malignant cells, suggesting the potential for anti-ganglioside antibody therapy for tumors. Anti-ganglioside GD2 antibody treatment is currently undergoing clinical trials for melanoma and neuroblastoma. We previously reported high in vivo antitumor effects of anti-GM2 ganglioside antibody against lung cancer. To determine whether anti-GM2 antibody may be clinically indicated for gastrointestinal cancers, we evaluated the mRNA expression of alpha2,8 sialyltransferase, a GD3 synthase, and beta1,4 N-acetylgalactosaminyltransferase (beta1,4 GalNAc-T), a GM2/GD2 synthase, in gastrointestinal cancers. We performed modified semi-quantitative RT-PCR, which reduces complexity incidental to radiolabeling on samples taken from small surgically removed clinical specimens. Stomach (19/22) and colorectal (21/30) cancers showed decreased expression of alpha2,8 sialyltransferase as compared with respective normal tissues (P < 0.05). In contrast, increased expression of beta1,4 GalNAc-T was detected in both types of tumors. Clinicopathological analysis revealed significantly higher expression level of alpha2,8 sialyltransferase in the poorly differentiated than in the well-differentiated stomach cancer group (P < 0.05). Furthermore, the expression level of alpha2,8 sialyltransferase was significantly decreased in male as compared with female colorectal cancer patients (P < 0.05). These results suggest that expression level of GM2 ganglioside is elevated in gastrointestinal cancer, and that anti-GM2 antibody may be applicable to its treatment.
Collapse
Affiliation(s)
- Yasuhiro Koh
- Pharmacology Division, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kramer K, Cheung NK. Antibody-based diagnostic and therapeutic innovations for human cancer. COMPREHENSIVE THERAPY 2002; 27:183-94. [PMID: 11569318 DOI: 10.1007/s12019-001-0013-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
As adjuvants, antibody-based diagnostic and therapeutic innovations can potentially decrease morbidity and mortality associated with many human malignancies. Current strategies employing genetically modified constructs may improve tumor penetration and increase versatility.
Collapse
Affiliation(s)
- K Kramer
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
16
|
Ren S, Kambe N, Du Z, Li Y, Xia HZ, Kambe M, Bieberich E, Pozez A, Grimes M, Yu RK, Irani AM, Schwartz LB. Disialoganglioside GD3 is selectively expressed by developing and mature human mast cells. J Allergy Clin Immunol 2001; 107:322-30. [PMID: 11174200 DOI: 10.1067/mai.2001.112272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Disialoganglioside GD3 is expressed on the surface of selected cell types. Anti-GD3 mAb administered to human subjects with malignant melanoma produces signs and symptoms of immediate hypersensitivity reactions. OBJECTIVE The expression of GD3 by human mast cells was assessed during mast cell development in vitro and in samples of lung and skin. METHODS GD3 on tissue- and in vitro-derived mast cells was analyzed after double labeling of cells for tryptase (G3 mAb) or Kit (YB5.B8 mAb) and GD3 (R24 mAb). Glycolipids in extracts of fetal liver-derived mast cells were examined by using high-performance thin-layer chromatography. RESULTS Flow cytometry showed that the percentage of GD3+ cells increased in parallel to Kit+ cells during the recombinant human stem cell factor-dependent development of fetal liver-derived mast cells. Double-labeling experiments showed that GD3+ cells were also surface Kit+ and granule tryptase positive, identifying them as mast cells in preparations of lung-, skin-, fetal liver-, and cord blood-derived cells. The major acidic glycolipid detected was NeuAcalpha2-8NeuAcalpha2-3Galbeta1-4Glcbeta1-1'Cer (GD3). Among peripheral blood leukocytes, only basophils and about 10% of the T cells were labeled with anti-GD3 mAb. Anti-GD3 mAb-conjugated magnetic beads were used to purify mast cells to greater than 90% purity from dispersed skin cells enriched to approximately 12% purity by means of density-dependent sedimentation but were less proficient for dispersed human lung mast cells, most likely because of other cell types that express GD3. CONCLUSION GD3 is expressed on the surface of developing human mast cells in parallel to tryptase in secretory granules and, like Kit, can serve as a target for their enrichment by immunoaffinity techniques.
Collapse
Affiliation(s)
- S Ren
- Department of Internal Medicine, Virginia Commonwealth University, Richmond 23298, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Coral S, Fonsatti E, Sigalotti L, De Nardo C, Visintin A, Nardi G, Colizzi F, Colombo MP, Romano G, Altomonte M, Maio M. Overexpression of protectin (CD59) down-modulates the susceptibility of human melanoma cells to homologous complement. J Cell Physiol 2000; 185:317-23. [PMID: 11056001 DOI: 10.1002/1097-4652(200012)185:3<317::aid-jcp1>3.0.co;2-l] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The clinical efficacy of therapeutic complement (C)-activating monoclonal antibodies (mAb) to melanoma-associated antigens can be impaired by the levels of expression of C-inhibitory molecules on neoplastic cells. Protectin (CD59) is a glycosylphosphatidylinositol (GPI)-anchored cell membrane glycoprotein, acting as terminal regulator of C cascade, which is heterogeneously expressed in melanomas and represents the main restriction factor of C-mediated lysis of melanoma cells. Thus, we investigated whether the overexpression of CD59 could influence the constitutive susceptibility of distinct melanoma cells to homologous C. Infection of CD59-positive Mel 100 and 70-W melanoma cells by a retroviral vector carrying the CD59 cDNA, significantly (P < 0.05) upregulated their constitutive expression of CD59, whereas it did not affect that of additional C-regulatory molecules. Transduced CD59 was entirely GPI-anchored and showed a molecular weight identical to native CD59. Additionally, higher amounts of soluble CD59 were detected in the conditioned media of CD59-transduced melanoma cells compared with parental cells. CD59-transduced melanoma cells, sensitized by the anti-GD3 disialoganglioside mAb R24, were significantly (P < 0.05) less susceptible to homologous C-lysis than were parental cells; this effect was fully reverted by the masking of CD59 with F(ab')(2) fragments of the anti-CD59 mAb YTH53.1. These results provide conclusive evidence demonstrating that absolute levels of CD59 expression regulate the susceptibility to homologous C of specific melanoma cells, and suggest an additional explanation for the poor clinical results obtained with C-activating mAb in the clinical setting.
Collapse
Affiliation(s)
- S Coral
- Advanced Immunotherapy Unit, Centro di Riferimento Oncologico, Istituto Nazionale di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zeng G, Gao L, Yu RK. Reduced cell migration, tumor growth and experimental metastasis of rat F-11 cells whose expression of GD3-synthase is suppressed. Int J Cancer 2000; 88:53-7. [PMID: 10962439 DOI: 10.1002/1097-0215(20001001)88:1<53::aid-ijc8>3.0.co;2-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We previously established a rat F-11 cell line whose expression of ganglioside GD3 was inhibited by stable transfection of the anti-sense vector against the GD3-synthase gene, showing that specific inhibition of GD3-synthase expression in tumor cells greatly reduced their growth rate in nude mice. Here, we report that down-regulation of GD3-synthase expression in anti-sense-transfected F-11 cells correlates with reduced cell migration and invasion in vitro and tumor growth and metastasis in vivo. When cultures were denuded of cells in a 1-mm-wide strip, the anti-sense-transfected F-11 cells migrated very slowly into the denuded area. Differences in migration between anti-sense-transfected cells and control parental cells were easily apparent. In vitro invasion assay of F-11 cells revealed a 3-fold decrease in invasion ability from the GD3-synthase-suppressed cells; colony formation in soft agar was not affected. Injection (i.v.) of control sense-transfected and untransfected F-11 cells resulted in multiple, large metastatic nodules in each of the 12 mice, whereas i.v. injection of anti-sense-transfected F-11 cells formed a single, small metastatic nodule in only 2 of the 8 nude mice. In addition, even if metastasis occurred, the anti-sense-induced metastatic nodules were much smaller than the metastatic nodules formed by control F-11 cells. These results demonstrate that suppression of GD3-synthase expression, which results primarily in a marked decrease in the concentration of ganglioside GD3, greatly reduces cell spreading, invasion and both the incidence and growth rate of experimental metastasis of F-11 cells.
Collapse
MESH Headings
- Animals
- Cell Division/physiology
- Cell Line, Transformed
- Cell Movement/physiology
- DNA, Antisense/genetics
- DNA, Antisense/pharmacology
- Female
- Ganglia, Spinal
- Gangliosides/biosynthesis
- Gangliosides/metabolism
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Male
- Mice
- Mice, Nude
- Neoplasm Metastasis
- Neoplasm Transplantation
- Neoplasms, Experimental/enzymology
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neuroblastoma
- Rats
- Sialyltransferases/antagonists & inhibitors
- Sialyltransferases/biosynthesis
- Sialyltransferases/genetics
- Transfection
Collapse
Affiliation(s)
- G Zeng
- Department of Biochemistry and Molecular Biophysics, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia, USA.
| | | | | |
Collapse
|
19
|
Sotelo J, Guevara P, Reyes S, Arrieta O. Interstitial quinacrine for elimination of abnormal tissue; therapy of experimental glioma. Surgery 2000; 128:439-46. [PMID: 10965316 DOI: 10.1067/msy.2000.107266] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND When quinacrine is injected interstitially, an intense migration of leukocytes and accumulation of various lymphokines is obtained locally, and the reaction is followed by cicatricial fibrosis. This property has been used in humans to induce tubal fibrosis in women and pleurodesis in patients with pleural effusion. METHODS In a controlled study, a single dose of 150 mg of quinacrine was injected interstitially into a C6 glioma implanted in the subcutaneous tissue of Wistar rats. Changes in size, histologic variations, and microscopic characteristics of leukocyte subpopulations infiltrating the tumor were studied by immunohistochemistry. Tumor necrosis factor and interleukin-1 beta were measured at different times in tumor homogenates. RESULTS The day after the injection of quinacrine, infiltration of leukocytes and macrophages was observed, accompanied by an accumulation of proinflammatory endogenous cytokines. Tumoral necrosis soon ensued; complete tumor disappearance was obtained in 72% of the animals. Cicatrization proceeded without injury of perilesional structures. In all controls injected with the vehicle, a large tumor developed (P <.0001). CONCLUSIONS Quinacrine, when administered interstitially in a single dose, elicits an intense local recruitment and proliferation of activated immune cells that, at the dose used in this study, induces tissue necrosis within a radius of 1 cm around the site of quinacrine injection, leaving the surrounding tissue unharmed.
Collapse
Affiliation(s)
- J Sotelo
- Neuroimmunology Unit, National Institute Of Neurology and Neurosurgery, Universidad Nacional Autonoma De Mexico
| | | | | | | |
Collapse
|
20
|
Fukumoto H, Nishio K, Ohta S, Hanai N, Fukuoka K, Ohe Y, Sugihara K, Kodama T, Saijo N. Effect of a chimeric anti-ganglioside GM2 antibody on ganglioside GM2-expressing human solid tumors in vivo. Int J Cancer 1999; 82:759-64. [PMID: 10417777 DOI: 10.1002/(sici)1097-0215(19990827)82:5<759::aid-ijc22>3.0.co;2-h] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ganglioside GM2 is expressed on the surface of neuroblastoma and glioblastoma cells, and may also be detected on lung cancer cells. We reported previously that anti-ganglioside GM2 antibody exhibited strong in vitro anti-tumor activity against adriamycin-resistant cancer cells, which overexpressed ganglioside GM2. In the present study, we examined the in vivo anti-tumor effect of the chimeric anti-ganglioside GM2 antibody, KM966, against human lung and breast carcinoma cells, SBC-3 and MCF-7, and respective adriamycin-resistant clones, SBC-3/ADM and AdrR MCF-7 in BALB/c nu/nu mice. Ratios of tumor volume (T/C) between KM966-treated group and control group were 0.01 for SBC-3, 0.00 for SBC-3/ADM, 0.85 for MCF-7 and 0.34 for AdrR MCF-7 cells, respectively. Nude mice, which were pretreated with anti-asialo GM1 antibody to remove natural killer cells, were transplanted with 4 x 10(7) of SBC-3 and SBC-3/ADM subcutaneously. Seven days later, when tumors had grown to a diameter of over 8 mm, mice began to receive intravenous treatment of 120 microgram/mouse KM966 daily. Fourteen daily treatments induced regression to less than 4-mm diameter in 4/5 SBC-3 tumors and 5/5 of SBC-3/ADM tumors. All SBC-3/ADM tumors disappeared completely, suggesting that KM966 exerts a strong in vivo anti-tumor effect on ganglioside GM2-expressing cancer cells. In KM966-treated mice, the surface of the tumor cells stained positive with anti-human IgG. In addition, numerous leukocytes had infiltrated into the tumor mass. Antibody-dependent cell-mediated cytotoxicity (ADCC) of KM966 against tumor cells was examined in vitro by (51)Cr-release assay and revealed that KM966 induces ADCC activity against ganglioside GM2-expressing tumors. Our results suggest that immunotherapy using KM966 may be useful for the treatment of ganglioside GM2-expressing solid tumors.
Collapse
Affiliation(s)
- H Fukumoto
- Pharmacology Division, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Weichenthal M, Siemann U, Neuber K, Breitbart EW. Expression of complement regulator proteins in primary and metastatic malignant melanoma. J Cutan Pathol 1999; 26:217-21. [PMID: 10408345 DOI: 10.1111/j.1600-0560.1999.tb01833.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The expression of complement regulatory antigens C3b/C4b receptor, (CD35) membrane cofactor protein (CD46), decay accelerating factor (CD55), and homologous restriction factor 20 (CD59) was determined immunohistochemically on ten primary malignant melanomas, 16 metastatic lesions, and ten melanocytic nevi. All of the melanocytic nevi and 9/10 primary melanomas showed both expression of CD46 and CD59. In one primary melanoma lacking CD46, expression of CD35 could be detected. In metastatic melanoma, 9/16 metastases were CD46+/CD59+, two were CD46-/CD59+, one CD46+/CD59-, and four CD46-/CD59-. Additionally, CD55 could be detected in two CD46+/CD59+ metastases, and CD35 in one. Expression or lack of complement regulatory antigens did not correlate with the expression of GD2, GD3, HMB-45 or S-100. In conclusion, some cases of metastatic melanoma show loss of normal expression of complement regulatory proteins. This might have implications on the immune response or the efficacy of immune therapy in malignant melanoma.
Collapse
Affiliation(s)
- M Weichenthal
- Department of Dermatology, St. Georg Hospital, Hamburg, Germany.
| | | | | | | |
Collapse
|
22
|
Qiu J, Luo P, Wasmund K, Steplewski Z, Kieber-Emmons T. Towards the development of peptide mimotopes of carbohydrate antigens as cancer vaccines. Hybridoma (Larchmt) 1999; 18:103-12. [PMID: 10211797 DOI: 10.1089/hyb.1999.18.103] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tumor-associated carbohydrate antigens are considered important targets in efforts to develop cancer vaccines. To further enhance vaccine efforts, we are developing peptide mimotopes of tumor-associated carbohydrate antigens that can elicit functional immune responses. Mapping peptide epitopes with anticarbohydrate antibodies can lend to defining structural relationships that can go undetected by screening of carbohydrate antigens alone. Here we contrast reactivity patterns for peptides using monoclonal antibodies (MAbs) directed to the neolactoseries related Lewis Y (LeY) and sialyl-Lewis X (sLeX) antigen and the GD3/GD2 ganglioside antigen. We observe that representative MAbs cross-react with a WRY-containing peptide and that this motif type is isolated by the respective monoclonal in peptide phage display screening. Primary immunization with multiple antigen peptide preparations with QS-21 adjuvant efficiently elicited cytotoxic IgM antibodies for a murine Meth A fibrosarcoma line expressing sLeX. The cytotoxicity of IgG polyclonal response was found to be as effective as IgM in mediating complement-dependent cytotoxicity against the Meth A line. These experiments suggest that peptide mimotopes of the LeY and sLeX tumor-associated carbohydrate antigen and QS-21 adjuvant could be considered as an immunogenic therapeutic vaccine in carcinoma and melanoma patients in the minimal residual disease setting.
Collapse
Affiliation(s)
- J Qiu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | |
Collapse
|
23
|
Alpaugh RK, von Mehren M, Palazzo I, Atkins MB, Sparano JA, Schuchter L, Weiner LM, Dutcher JP. Phase IB trial for malignant melanoma using R24 monoclonal antibody, interleukin-2/alpha-interferon. Med Oncol 1998; 15:191-8. [PMID: 9819796 DOI: 10.1007/bf02821938] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The inflammatory tumor lymphocytic infiltrates and spontaneous tumor regressions seen in patients with metastatic malignant melanomas suggest a cellular immune involvement. Enhancement of such responses has been the goal of R24 (GD3 ganglioside-specific) monoclonal antibody trials, alone and in combination with other agents. This study reports the results of 21 patients treated in a phase IB trial employing R24 (0, 5, 25, 50 mg/m2) administered by continuous i.v. infusion on days 1-5 followed by 3 MU each of interleukin-2 (IL-2) and alpha interferon (alpha-IFN) given subcutaneously on days 8-12, 15-19 and 22-26. R24-related toxicities occurred pre-dominantly at the 25 and 50 mg/m2 doses. One patient (50 mg/m2 R24) exhibited a dose-limiting Grade 4 anaphylaxis. Cytokine-related toxicities required IL-2/alpha-IFN dose reduction in two patients and early termination of treatment in five additional patients. Nine of 20 baseline biopsies showed chronic inflammation; six with lymphocytic tumor infiltration and three where inflammation was confined to the perivascular/peritumoral spaces. No day 8 or 29 biopsies in the R24-treated groups demonstrated treatment-induced tumor lymphocytic infiltrates. However, one patient randomized to no R24 treatment, showed a significant inflammatory tumor lymphocytic infiltration at days 8 and 29. Eighteen of 21 treated patients were evaluable for response. One (5%) patient receiving IL-2/alpha-IFN alone had stable disease lasting 1.5 years. Five (28%) R24, IL-2/alpha-IFN-treated patients had stable disease ranging from 6 to 32 weeks, with one patient remaining alive 2.5 years post-treatment. Although this combined treatment program was generally well tolerated, no objective responses were seen and significant R24-induced tumor lymphocytic infiltrates were not demonstrated.
Collapse
Affiliation(s)
- R K Alpaugh
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Maguire HC, Berd D, Lattime EC, McCue PA, Kim S, Chapman PB, Mastrangelo MJ. Phase I study of R24 in patients with metastatic melanoma including evaluation of immunologic parameters. Cancer Biother Radiopharm 1998; 13:13-23. [PMID: 10850338 DOI: 10.1089/cbr.1998.13.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
R24 is a mouse IgG3 monoclonal antibody with specificity for the disialoganglioside GD3. Most human melanomas have substantial surface GD3; in addition, a significant proportion of T lymphocytes display surface GD3. In a phase I study, we have investigated the toxicity and effect on selected immunological parameters of three dose levels of R24 given intravenously daily for five days (10 mg/m2/d, 30 mg/m2/d and 50 mg/m2/d) to patients with advanced melanoma. R24 administration neither consistently diminished nor augmented expression of delayed type hypersensitivity (DTH) skin reactions to anergy panel antigens or to a contact allergen dinitrofluorobenzene. R24 was infrequently found on tumor cells, or on lymphocytes from DTH biopsies, despite measurable serum levels of R24. The 30 mg/m2/d dose of R24 produced a statistically significant drop in peripheral blood lymphocytes on treatment Day 5. Likewise, on Day 5 there was a modest but statistically significant decrement in the proportion of circulating cells which were R24+. While there was one mixed response, there were no complete or partial tumor regressions in the R24 treated patients; there was no evident clinical benefit from the R24 therapy. The toxicity of the R24 at the higher dose levels can be very substantial. One patient, on the highest dose level, died on the 4th day of R24 treatment; in the absence of a plausible alternative explanation, a relationship of the death to the administered R24 must be considered. A precipitous drop in serum albumin coincident with R24 administration was found in all cases; this effect has not been previously reported with R24.
Collapse
Affiliation(s)
- H C Maguire
- Department of Medicine (Division of Medical Oncology), Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Clinical research in the area of antibody-based tumor-targeted therapy has been driven for many years by the prospect of identifying cell surface antigens with sufficient restrictive tissue expression patterns to allow for the selective and specific accumulation of antibody in tumor tissue. Few, if any, such antibody-antigen systems have been identified which can effectively deliver a large fraction of an administered therapeutic agent to metastatic cancer. Despite this limitation, however, a greater understanding of the biological and physiological principles of tumor-targeted therapy has resulted in successful antibody-based therapy of lymphoma, colon cancer and breast cancer in recent clinical trials.
Collapse
Affiliation(s)
- A M Scott
- Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch, Austin, Australia.
| | | |
Collapse
|
26
|
Hartmann F, Pfreundschuh M. [Oncology '96]. MEDIZINISCHE KLINIK (MUNICH, GERMANY : 1983) 1997; 92:83-100. [PMID: 9139216 DOI: 10.1007/bf03042290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- F Hartmann
- Medizinische Klinik und Poliklinik, Universität des Saarlandes, Homburg (Saar)
| | | |
Collapse
|
27
|
Gruber R, Holz E, Riethmüller G. Monoclonal antibodies in cancer therapy. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1996; 18:243-51. [PMID: 8908703 DOI: 10.1007/bf00820669] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- R Gruber
- Institut für Immunologle, Ludwig Maximilians-Universitt, München, Germany
| | | | | |
Collapse
|