1
|
Hereditary angioedema caused by c1-esterase inhibitor deficiency: a literature-based analysis and clinical commentary on prophylaxis treatment strategies. World Allergy Organ J 2013; 4:S9-S21. [PMID: 23283143 PMCID: PMC3666183 DOI: 10.1097/wox.0b013e31821359a2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hereditary angioedema (HAE) caused by C1-esterase inhibitor deficiency is an autosomal-dominant disease resulting from a mutation in the C1-inhibitor gene. HAE is characterized by recurrent attacks of intense, massive, localized subcutaneous edema involving the extremities, genitalia, face, or trunk, or submucosal edema of upper airway or bowels. These symptoms may be disabling, have a dramatic impact on quality of life, and can be life-threatening when affecting the upper airways. Because the manifestations and severity of HAE are highly variable and unpredictable, patients need individualized care to reduce the burden of HAE on daily life. Although effective therapy for the treatment of HAE attacks has been available in many countries for more than 30 years, until recently, there were no agents approved in the United States to treat HAE acutely. Therefore, prophylactic therapy is an integral part of HAE treatment in the United States and for selected patients worldwide. Routine long-term prophylaxis with either attenuated androgens or C1-esterase inhibitor has been shown to reduce the frequency and severity of HAE attacks. Therapy with attenuated androgens, a mainstay of treatment in the past, has been marked by concern about potential adverse effects. C1-esterase inhibitor works directly on the complement and contact plasma cascades to reduce bradykinin release, which is the primary pathologic mechanism in HAE. Different approaches to long-term prophylactic therapy can be used to successfully manage HAE when tailored to meet the needs of the individual patient.
Collapse
|
2
|
Illidge T, Morschhauser F. Radioimmunotherapy in follicular lymphoma. Best Pract Res Clin Haematol 2011; 24:279-93. [DOI: 10.1016/j.beha.2011.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
3
|
Ivanov A, Swann R, Illidge T. New insights into the mechanisms of action of radioimmunotherapy in lymphoma. J Pharm Pharmacol 2008; 60:987-98. [PMID: 18644192 DOI: 10.1211/jpp.60.8.0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The exquisite sensitivity of haematological malignancies to targeted radiation alongside the impressive results achieved by the pioneers in this field suggests that radioimmunotherapy is likely to be a productive area for future clinical research. Recent experimental work has demonstrated that the combination of targeted radiation and antibody effector mechanisms are critical to long-term clearance of tumour. This review provides the background of clinical and biological insights into the mechanisms of action of radioimmunotherapy.
Collapse
Affiliation(s)
- Andrei Ivanov
- School of Cancer and Imaging Sciences, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
4
|
Walker JA, Smith KGC. Dependence of surface monoclonal antibody binding on dynamic changes in FcgammaRIIb expression. Immunology 2008; 124:412-8. [PMID: 18217946 PMCID: PMC2440835 DOI: 10.1111/j.1365-2567.2007.02791.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Receptors for the Fc region of immunoglobulin G (FcgammaRs) are expressed on a broad range of haematopoietic cell types and are responsible for regulating antibody production and linking the humoral and effector responses. In response to a number of stimuli, such as cytokine signals or inflammation, FcgammaR expression at the cell surface is dynamically regulated. On B cells, we observed what appeared to be a correlation between CD22 expression and FcgammaRIIb expression when the latter was varied in a number of models. Further investigation revealed that this was specific to a particular anti-CD22 monoclonal antibody, which appeared to require stabilization by interaction with FcgammaRIIb for optimal binding to CD22. Since alterations in the regulation of FcgammaR expression are important in controlling immune responses and have been associated with a number of immune-mediated disease states, we suggest that it might be prudent to confirm the expression of cell surface markers by two independent methods. Furthermore, because the efficacy of therapeutic antibodies may depend upon their interaction with FcgammaRs, our results are relevant to their design and assessment.
Collapse
Affiliation(s)
- Jennifer A Walker
- Department of Medicine, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, UK.
| | | |
Collapse
|
5
|
Brans B, Bodei L, Giammarile F, Linden O, Luster M, Oyen WJG, Tennvall J. Clinical radionuclide therapy dosimetry: the quest for the "Holy Gray". Eur J Nucl Med Mol Imaging 2007; 34:772-786. [PMID: 17268773 PMCID: PMC1914264 DOI: 10.1007/s00259-006-0338-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction Radionuclide therapy has distinct similarities to, but also profound differences from external radiotherapy. Review This review discusses techniques and results of previously developed dosimetry methods in thyroid carcinoma, neuro-endocrine tumours, solid tumours and lymphoma. In each case, emphasis is placed on the level of evidence and practical applicability. Although dosimetry has been of enormous value in the preclinical phase of radiopharmaceutical development, its clinical use to optimise administered activity on an individual patient basis has been less evident. In phase I and II trials, dosimetry may be considered an inherent part of therapy to establish the maximum tolerated dose and dose-response relationship. To prove that dosimetry-based radionuclide therapy is of additional benefit over fixed dosing or dosing per kilogram body weight, prospective randomised phase III trials with appropriate end points have to be undertaken. Data in the literature which underscore the potential of dosimetry to avoid under- and overdosing and to standardise radionuclide therapy methods internationally are very scarce. Developments In each section, particular developments and insights into these therapies are related to opportunities for dosimetry. The recent developments in PET and PET/CT imaging, including micro-devices for animal research, and molecular medicine provide major challenges for innovative therapy and dosimetry techniques. Furthermore, the increasing scientific interest in the radiobiological features specific to radionuclide therapy will advance our ability to administer this treatment modality optimally.
Collapse
Affiliation(s)
- B Brans
- Department of Nuclear Medicine, University Hospital Maastricht, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - L Bodei
- Division of Nuclear Medicine, Istituto Europeo di Oncologia, Milan, Italy
| | - F Giammarile
- Service de Médecine Nucléaire, Centre Léon Bérard, Lyon, France
| | - O Linden
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - M Luster
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - W J G Oyen
- Department of Nuclear Medicine, University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - J Tennvall
- Department of Oncology, Lund University Hospital, Lund, Sweden
| |
Collapse
|
6
|
Corneillie TM, Whetstone PA, Meares CF. Irreversibly binding anti-metal chelate antibodies: Artificial receptors for pretargeting. J Inorg Biochem 2006; 100:882-90. [PMID: 16487590 DOI: 10.1016/j.jinorgbio.2006.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 12/14/2005] [Accepted: 01/02/2006] [Indexed: 10/25/2022]
Abstract
Antibodies against metal chelates may potentially be used in biomedical applications such as targeted imaging and therapy of cancer. Highly specific monoclonal antibodies can be developed, but their binding strength needs to be maximized for them to be of practical use. In general, the half-life for dissociation of an antibody-ligand complex is more than an order of magnitude lower than the half-lifetimes for decay of medically useful radiometal ions. Practically speaking, the metal chelate-based ligand will not be bound to its receptor long enough for all of the bound radiometal to decay. A novel approach to this problem is a combination of synthetic chemistry and site-directed mutagenesis, to position a mildly reactive group on the metal chelate adjacent to a complementary reactive group on the antibody when the complex is formed. The partners are chosen to be sufficiently unreactive so that they coexist with other molecules in living systems without undergoing reaction. When the antibody-chelate complex is formed the effective local concentrations of the two groups can be non-physically large, so that a permanent link is formed in the complex even though no reaction occurs when the partners are free in solution.
Collapse
Affiliation(s)
- Todd M Corneillie
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA
| | | | | |
Collapse
|
7
|
Vose JM. Bexxar
®
: Novel Radioimmunotherapy for the Treatment of Low‐Grade and Transformed Low‐Grade Non‐Hodgkin's Lymphoma. Oncologist 2004; 9:160-72. [PMID: 15047920 DOI: 10.1634/theoncologist.9-2-160] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Immunotherapy using monoclonal antibodies to specifically target B cells has provided new hope to many patients with indolent lymphomas, particularly those with chemotherapy-refractory disease. Lymphomas are extremely sensitive to radiation, and significant progress has been made over the last decade in the development of radioimmunotherapy with anti-CD20 antibodies. MATERIALS AND METHODS Herein we review clinical experience with tositumomab and iodine I 131 tositumomab (Bexxar; Corixa Corporation; South San Francisco, CA; and GlaxoSmithKline; Philadelphia, PA) in patients with non-Hodgkin's lymphoma. RESULTS Therapy with Bexxar has demonstrated high response rates and long durations of response compared with unconjugated anti-CD20 antibodies in patients with relapsed low-grade and transformed low-grade non-Hodgkin's lymphomas. Iodine-131 (I-131) has a long history of clinical experience, an excellent safety record, and favorable nuclear and pharmacologic properties. Importantly, the gamma emissions of iodine-131 facilitate accurate dosimetry to calculate the appropriate patient-specific therapeutic activity to deliver a predetermined total-body dose of radiation, thereby minimizing hematologic toxicity. In clinical trials of Bexxar, objective response rates ranged from 54%-71% in heavily pretreated patients. In the pivotal trial, the number of patients with a longer duration of response after treatment with Bexxar was significantly greater than the number of patients with a longer duration of response after their last qualifying chemotherapy regimen. In 76 newly diagnosed patients, the objective response rate was 97%, and 63% of patients achieved complete responses. CONCLUSION These data suggest that Bexxar will become an important new option in the treatment of indolent lymphoma.
Collapse
Affiliation(s)
- Julie M Vose
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198-7680, USA.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Pontiggia L, Lassila R, Pederiva S, Schmid HR, Burger M, Beer JH. Increased platelet-collagen interaction associated with double homozygosity for receptor polymorphisms of platelet GPIa and GPIIIa. Arterioscler Thromb Vasc Biol 2002; 22:2093-8. [PMID: 12482840 DOI: 10.1161/01.atv.0000042230.26207.d2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE There is considerable controversy regarding the clinical role of the single-nucleotide polymorphisms (SNPs) of the platelet glycoprotein receptor GPIa C807T and the Pl(A1/A2) of GPIIIa as cardiovascular risk factors. We hypothesized that two combined SNPs in their homozygous prothrombotic forms could clarify their pathophysiological impact. METHODS AND RESULTS We identified a family with a striking history of premature cardiovascular events and a high frequency of the prothrombotic form of the two SNPs. From this family, the platelets of a healthy, 27-year-old propositus with this double homozygosity were compared with three matched male neutral gene variant controls. The propositus had shortened PFA-100 closure times and an increased platelet aggregation response to collagen. Platelet deposition to collagen was augmented under the blood flow conditions of a high shear rate model (1600 s(-1)). Platelet adhesion on collagen monomers was induced in a static system, leading to the promotion of subsequent procoagulant activity. CONCLUSIONS The combined homozygous prothrombotic SNPs of GPIa and GPIIIa are associated with an increased platelet-collagen interaction and procoagulant activity that can be readily demonstrated in several independent systems. Our patient may serve as a useful model for the functional consequences of two combined, potentially procoagulant, platelet SNPs.
Collapse
Affiliation(s)
- Luca Pontiggia
- Department of Medicine, Laboratory for Thrombosis Research, Kantonsspital Baden, Switzerland
| | | | | | | | | | | |
Collapse
|
11
|
Kosmas C, Stamatopoulos K, Stavroyianni N, Tsavaris N, Papadaki T. Anti-CD20-based therapy of B cell lymphoma: state of the art. Leukemia 2002; 16:2004-15. [PMID: 12357351 DOI: 10.1038/sj.leu.2402639] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2002] [Accepted: 05/07/2002] [Indexed: 11/09/2022]
Abstract
Over the last 5 years, studies applying the chimeric anti-CD20 MAb have renewed enthusiasm and triggered world-wide application of anti-CD20 MAb-based therapies in B cell non-Hodgkin's lymphoma (NHL). Native chimeric anti-CD20 and isotope-labeled murine anti-CD20 MAbs are currently employed with encouraging results as monotherapy or in combination with conventional chemotherapy and in consolidation of remission after treatments with curative intent (ie after/ in combination with high-dose chemotherapy and hematopoietic stem cell rescue). On the available experience, anti-CD20 MAb-based therapeutic strategies will be increasingly integrated in the treatment of B cell NHL and related malignancies.
Collapse
Affiliation(s)
- C Kosmas
- Department of Medicine, 2nd Division of Medical Oncology, Metaxa Cancer Hospital, Piraeus, Greece
| | | | | | | | | |
Collapse
|
12
|
Benvenuti F, Burrone OR. Anti-idiotypic antibodies induced by genetic immunisation are directed exclusively against combined V(L)/V(H) determinants. Gene Ther 2001; 8:1555-61. [PMID: 11704816 DOI: 10.1038/sj.gt.3301546] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2001] [Accepted: 07/12/2001] [Indexed: 11/09/2022]
Abstract
DNA vaccines encoding the idiotype of immunoglobulins of tumour B cells were shown to induce protection in several mouse lymphoma models. The mechanism of rejection of tumour cells has not been fully understood, but there is strong evidence suggesting that engagement of the idiotype by anti-idiotypic antibodies may directly result in inhibition of tumour growth. In this study, we have investigated the structural basis of the idiotypic/anti-idiotypic interaction following immunisation with DNA vaccines. scFvs containing only one of the two tumour-derived V regions recombined to an irrelevant V region partner were generated. These constructs encoding a secretory form of the scFv were used as immunogens to induce anti-Id antibodies. The same scFvs were expressed as membrane-bound molecules on the surface of mammalian cells. Analysis of immune sera on the membrane-displayed idiotypes revealed that DNA immunisation induced a polyclonal antibody response restricted to conformational combined epitopes formed by the parental V(L)/V(H) association. Immune sera raised by scFv DNA vaccination did not show any detectable reactivity towards chimeric scFvs containing only one of the two immunising V regions, indicating that the response against combined V(L)/V(H)determinants is highly dominant. Remarkably, the same immunogen, delivered as scFv protein, induced antibodies also directed against chain-specific determinants. These findings indicate that presentation of properly folded idiotypes results in a highly specific antibody response directed exclusively to private idiotypic determinants of the V(L)/V(H) combination of the immunogen.
Collapse
Affiliation(s)
- F Benvenuti
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34012 Trieste, Italy
| | | |
Collapse
|
13
|
Wun T, Kwon DS, Tuscano JM. Radioimmunotherapy: potential as a therapeutic strategy in non-Hodgkin's lymphoma. BioDrugs 2001; 15:151-62. [PMID: 11437681 DOI: 10.2165/00063030-200115030-00002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Lymphomas are the fifth most common malignancy in the United States and are increasing in incidence. Despite being among the most responsive malignancies to radiation and chemotherapy, the majority of patients relapse or have progressive disease. Monoclonal antibodies (MAbs) directed at cell-specific surface antigens have been useful in the diagnosis of lymphomas and, more recently, the therapeutic mouse-human chimeric MAb rituximab has demonstrated effectiveness in B cell lymphomas. Conjugating MAbs to radionuclides is a strategy for improving the efficacy of MAb lymphoma therapy by delivering radiation in close proximity to the tumour (radioimmunotherapy or RIT). In addition, the low dose rate of the delivered radiation may exert a greater antitumour activity than an equivalent dose of conventional external beam radiation. The antigenic targets for MAb therapy have included CD20, CD22, HLA-DR, and B cell idiotype. Radionuclides that have been used include iodine-131, yttrium-90, and copper-67; there are relative merits and disadvantages to each source of radiation. Clinical studies to date have focused on relapsed and refractory patients with both indolent and aggressive lymphomas, although more recent studies have included previously untreated patients with indolent lymphoma. Radioimmunoconjugate has been delivered as either single or multiple doses. Response rates have varied widely, dependent on the patient population and the response criteria. Of note, complete responses can be achieved in this typically refractory patient group. Toxicities have generally consisted of mild infusion-related nausea, fever, chills, and asthenia. Neutropenia and thrombocytopenia are the dose-limiting toxicities and have prompted the incorporation of autologous stem cell support as a means of achieving dose escalation. To date, RIT has been delivered to highly selected patients in relatively few centres with requisite equipment and specialised personnel. In addition to these requirements, cost is likely to be a barrier to widespread use. The combination of RIT with chemotherapy at conventional or high dose, or with biological agents is a fertile area for investigation. The potential of RIT in the treatment for lymphomas will be defined only by well designed comparative prospective clinical studies.
Collapse
Affiliation(s)
- T Wun
- Division of Hematology Oncology, University of Calirnia Davis School of Medicine, Sacramento, California, USA.
| | | | | |
Collapse
|
14
|
Abstract
The idea of using the specificity of antibodies to target malignant cells was put forward very soon after the discovery of techniques to generate monoclonal reagents. The responses seen with mouse anti-idiotype in patients with B-cell lymphomas indicated the potential of this approach, but it was some years before key technical obstacles were overcome and the more widespread application of these therapies became possible. Whilst they were originally conceived as having an immunotherapeutic effect, it has become clear that recruitment of immune effectors is only one component of successful antibody therapy, and their action upon the cellular target, either blocking or agonistic, is also critical. The development of immunoconjugates to deliver toxins or radiation is a further extension of the approach, and here again the intracellular effect of antibody ligation appears to be crucial. This presentation will address the central theme of antibody treatments for malignancy that are now reaching the clinic, and will use these examples to highlight ways in which antibodies may be acting in vivo.
Collapse
MESH Headings
- Abatacept
- Adjuvants, Immunologic/therapeutic use
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/genetics
- Antibodies, Neoplasm/therapeutic use
- Antibody-Dependent Cell Cytotoxicity
- Antigens, CD
- Antigens, Differentiation/immunology
- Apoptosis
- Biological Availability
- CTLA-4 Antigen
- Half-Life
- Humans
- Immunization, Passive
- Immunoconjugates
- Neoplasms/immunology
- Neoplasms/therapy
- Protein Engineering
- Receptors, Growth Factor/antagonists & inhibitors
- Trastuzumab
Collapse
Affiliation(s)
- P W Johnson
- Cancer Sciences Division, Southampton University of School of Medicine, Southampton General Hospital, United Kingdom.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Abstract
Much of the 25 years since Kohler and Milstein first described making monoclonal antibodies (mAbs) has been spent trying to develop these reagents to treat human disease. Until recently, progress has been frustratingly slow and by 1994 only one mAb, anti-CD3 (OKT3), had been licensed for clinical use. In the past five years, however, the situation has changed dramatically, with numerous mAbs now showing clinical potential, and a further seven approved for human treatment. Furthermore, all indications are that this upward trend will continue, with a quarter of all new biological products currently undergoing clinical development being antibody based.
Collapse
Affiliation(s)
- M J Glennie
- Tenovus Research Laboratory, The Cancer Sciences Division, Southampton University School of Medicine, General Hospital, Southampton, UK SO16 6YD.
| | | |
Collapse
|
18
|
Abstract
Experimental radioimmunotherapy (RIT) studies in animal models have contributed significantly to the design of clinical RIT protocols, although the results have not always been directly translated. Reviewed in this article are current areas of active research in experimental RIT to increase the therapeutic ratio that are likely to have a significant impact on the design of future clinical studies. Approaches for increasing the therapeutic efficacy of RIT include the development of new targeting molecules (genetically engineered monoclonal antibodies, antibody fragments, single-chain antibodies, diabodies and minibodies, fusion toxins, or peptides); improved labeling chemistry; novel radionuclide use and fractionation; locoregional administration; pretargeting; use of biological response modifiers or gene transfer techniques to increase target receptor expression; bone marrow transplantation; and combined modality therapy with external-beam radiation therapy, chemotherapy, or gene therapy. Further research with these new experimental approaches in preclinical animal models is necessary to contribute to advances in the treatment of cancer patients using radiolabeled antibodies and peptides.
Collapse
Affiliation(s)
- D J Buchsbaum
- Division of Radiation Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
19
|
Affiliation(s)
- T M Illidge
- CRC Oncology Unit, Cancer Sciences Division, Southampton University School of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK
| | | |
Collapse
|
20
|
Abstract
Over the past 10-15 years, genetic engineering of monoclonal antibodies has greatly improved their utility in humans and in particular their ability to recruit immunological effectors such as natural killer cells and macrophages. Clinical results now confirm that these new reagents, when directed at the appropriate tumor markers (e.g. CD20 or Her-2), can control disease without untoward side effects. However, despite such success it is still unclear exactly how monoclonal antibodies (mAbs) destroy tumors in vivo. The ability of mAbs to crosslink membrane receptors and generate intracellular signals is part of the mechanism by which they control tumor growth. New data show that such 'signaling' mAbs can be used to sensitize tumors to the action of conventional DNA-damaging drugs.
Collapse
Affiliation(s)
- M S Cragg
- Cancer Sciences Division, Tenovus Research Laboratory, General Hospital, Southampton, SO16 6YD, UK.
| | | | | |
Collapse
|