1
|
Hegde M, Girisa S, Aswani BS, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Harnessing potential role of gangliosides in immunomodulation and cancer therapeutics. Life Sci 2024; 351:122786. [PMID: 38848944 DOI: 10.1016/j.lfs.2024.122786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/01/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Gangliosides represent glycolipids containing sialic acid residues, present on the cell membrane with glycan residues exposed to the extracellular matrix (ECM), while the ceramides are anchored within the membrane. These molecules play a critical role in pathophysiological processes such as host-pathogen interactions, cell-cell recognition, signal transduction, cell adhesion, motility, and immunomodulation. Accumulated evidence suggests the overexpression of gangliosides on tumor tissues in comparison to healthy human tissues. These tumor-associated gangliosides have been implicated in various facets of tumor biology, including cell motility, differentiation, signaling, immunosuppression, angiogenesis, and metastasis. Consequently, these entities emerge as attractive targets for immunotherapeutic interventions. Notably, the administration of antibodies targeting gangliosides has demonstrated cytotoxic effects on cancer cells that exhibit an overexpression of these glycolipids. Passive immunotherapy approaches utilizing murine or murine/human chimeric anti-ganglioside antibodies have been explored as potential treatments for diverse cancer types. Additionally, vaccination strategies employing tumor-associated gangliosides in conjunction with adjuvants have entered the realm of promising techniques currently undergoing clinical trials. The present comprehensive review encapsulates the multifaceted roles of gangliosides in tumor initiation, progression, immunosuppression, and metastasis. Further, an overview is provided of the correlation between the expression status of gangliosides in normal and tumor cells and its impact on cancer patient survival. Furthermore, the discussion extends to ongoing and completed clinical trials employing diverse strategies to target gangliosides, elucidating their effectiveness in treating cancers. This emerging discipline is expected to supply substantial impetus for the establishment of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India.
| |
Collapse
|
2
|
Homogeneous antibody and CAR-T cells with improved effector functions targeting SSEA-4 glycan on pancreatic cancer. Proc Natl Acad Sci U S A 2021; 118:2114774118. [PMID: 34876527 DOI: 10.1073/pnas.2114774118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2021] [Indexed: 01/07/2023] Open
Abstract
Pancreatic cancer is usually asymptomatic in the early stages; the 5-y survival rate is around 9%; and there is a lack of effective treatment. Here we show that SSEA-4 is more expressed in all pancreatic cancer cell lines examined but not detectable in normal pancreatic cells; and high expression of SSEA-4 or the key enzymes B3GALT5 + ST3GAL2 associated with SSEA-4 biosynthesis significantly lowers the overall survival rate. To evaluate potential new treatments for pancreatic cancer, homogeneous antibodies with a well-defined Fc glycan for optimal effector functions and CAR-T cells with scFv construct designed to target SSEA-4 were shown highly effective against pancreatic cancer in vitro and in vivo. This was further supported by the finding that a subpopulation of natural killer (NK) cells isolated by the homogeneous antibody exhibited enhancement in cancer-cell killing activity compared to the unseparated NK cells. These results indicate that targeting SSEA-4 by homologous antibodies or CAR-T strategies can effectively inhibit cancer growth, suggesting SSEA-4 as a potential immunotherapy target for treating pancreatic disease.
Collapse
|
3
|
Thomas A, Sumughan S, Dellacecca ER, Shivde RS, Lancki N, Mukhatayev Z, Vaca CC, Han F, Barse L, Henning SW, Zamora-Pineda J, Akhtar S, Gupta N, Zahid JO, Zack SR, Ramesh P, Jaishankar D, Lo AS, Moss J, Picken MM, Darling TN, Scholtens DM, Dilling DF, Junghans RP, Le Poole IC. Benign tumors in TSC are amenable to treatment by GD3 CAR T cells in mice. JCI Insight 2021; 6:e152014. [PMID: 34806651 PMCID: PMC8663788 DOI: 10.1172/jci.insight.152014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/14/2021] [Indexed: 11/23/2022] Open
Abstract
Mutations underlying disease in tuberous sclerosis complex (TSC) give rise to tumors with biallelic mutations in TSC1 or TSC2 and hyperactive mammalian target of rapamycin complex 1 (mTORC1). Benign tumors might exhibit de novo expression of immunogens, targetable by immunotherapy. As tumors may rely on ganglioside D3 (GD3) expression for mTORC1 activation and growth, we compared GD3 expression in tissues from patients with TSC and controls. GD3 was overexpressed in affected tissues from patients with TSC and also in aging Tsc2+/- mice. As GD3 overexpression was not accompanied by marked natural immune responses to the target molecule, we performed preclinical studies with GD3 chimeric antigen receptor (CAR) T cells. Polyfunctional CAR T cells were cytotoxic toward GD3-overexpressing targets. In mice challenged with Tsc2-/- tumor cells, CAR T cells substantially and durably reduced the tumor burden, correlating with increased T cell infiltration. We also treated aged Tsc2+/- heterozygous (>60 weeks) mice that carry spontaneous Tsc2-/- tumors with GD3 CAR or untransduced T cells and evaluated them at endpoint. Following CAR T cell treatment, the majority of mice were tumor free while all control animals carried tumors. The outcomes demonstrate a strong treatment effect and suggest that targeting GD3 can be successful in TSC.
Collapse
Affiliation(s)
- Ancy Thomas
- Department of Dermatology, Feinberg School of Medicine
- Robert H. Lurie Comprehensive Cancer Center
| | | | | | | | - Nicola Lancki
- Quantitative Data Sciences Core, Robert H. Lurie Comprehensive Cancer Center; and
| | | | | | - Fei Han
- Department of Dermatology, Feinberg School of Medicine
- Robert H. Lurie Comprehensive Cancer Center
| | - Levi Barse
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Jesus Zamora-Pineda
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Suhail Akhtar
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Nikhilesh Gupta
- Robert H. Lurie Comprehensive Cancer Center
- Illinois Mathematics and Science Academy, Aurora, Illinois, USA
| | - Jasmine O. Zahid
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Stephanie R. Zack
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | | | | | - Agnes S.Y. Lo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Maria M. Picken
- Department of Pathology, Loyola University, Maywood, Illinois, USA
| | - Thomas N. Darling
- Department of Dermatology, School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Denise M. Scholtens
- Quantitative Data Sciences Core, Robert H. Lurie Comprehensive Cancer Center; and
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniel F. Dilling
- Department of Medicine, Stritch School of Medicine, Loyola University, Maywood, Illinois, USA
| | - Richard P. Junghans
- Department of Hematology/Oncology, School of Medicine, Boston University, Boston, Massachusetts, USA
| | - I. Caroline Le Poole
- Department of Dermatology, Feinberg School of Medicine
- Robert H. Lurie Comprehensive Cancer Center
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
4
|
Cancer-Associated Glycosphingolipids as Tumor Markers and Targets for Cancer Immunotherapy. Int J Mol Sci 2021; 22:ijms22116145. [PMID: 34200284 PMCID: PMC8201009 DOI: 10.3390/ijms22116145] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Aberrant expression of glycosphingolipids is a hallmark of cancer cells and is associated with their malignant properties. Disialylated gangliosides GD2 and GD3 are considered as markers of neuroectoderm origin in tumors, whereas fucosyl-GM1 is expressed in very few normal tissues but overexpressed in a variety of cancers, especially in small cell lung carcinoma. These gangliosides are absent in most normal adult tissues, making them targets of interest in immuno-oncology. Passive and active immunotherapy strategies have been developed, and have shown promising results in clinical trials. In this review, we summarized the current knowledge on GD2, GD3, and fucosyl-GM1 expression in health and cancer, their biosynthesis pathways in the Golgi apparatus, and their biological roles. We described how their overexpression can affect intracellular signaling pathways, increasing the malignant phenotypes of cancer cells, including their metastatic potential and invasiveness. Finally, the different strategies used to target these tumor-associated gangliosides for immunotherapy were discussed, including the use and development of monoclonal antibodies, vaccines, immune system modulators, and immune effector-cell therapy, with a special focus on adoptive cellular therapy with T cells engineered to express chimeric antigen receptors.
Collapse
|
5
|
Ghosh S, Trabbic KR, Shi M, Nishat S, Eradi P, Kleski KA, Andreana PR. Chemical synthesis and immunological evaluation of entirely carbohydrate conjugate Globo H-PS A1. Chem Sci 2020; 11:13052-13059. [PMID: 34123241 PMCID: PMC8163331 DOI: 10.1039/d0sc04595k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022] Open
Abstract
An anticancer, entirely carbohydrate conjugate, Globo H-polysaccharide A1 (Globo H-PS A1), was chemically prepared and immunologically evaluated in C57BL/6 mice. Tumor associated carbohydrate antigen Globo H hexasaccharide was synthesized in an overall 7.8% yield employing a convergent [3 + 3] strategy that revealed an anomeric aminooxy group used for conjugation to oxidized PS A1 via an oxime linkage. Globo H-PS A1, formulated with adjuvants monophosphoryl lipid A and TiterMax® Gold. After immunization an antigen specific immune response was observed in ELISA with anti-Globo H IgG/IgM antibodies. Specificity of the corresponding antibodies was determined by FACS showing cell surface binding to Globo H-positive cancer cell lines MCF-7 and OVCAR-5. The anti-Globo H antibodies also exhibited complement-dependent cellular cytotoxicity against MCF-7 and OVCAR-5 cells.
Collapse
Affiliation(s)
- Samir Ghosh
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Kevin R Trabbic
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Mengchao Shi
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Sharmeen Nishat
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Pradheep Eradi
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Kristopher A Kleski
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| | - Peter R Andreana
- The University of Toledo, Department of Chemistry and Biochemistry 2801 West Bancroft Street Toledo Ohio USA 43606
| |
Collapse
|
6
|
Cox EC, Thornlow DN, Jones MA, Fuller JL, Merritt JH, Paszek MJ, Alabi CA, DeLisa MP. Antibody-Mediated Endocytosis of Polysialic Acid Enables Intracellular Delivery and Cytotoxicity of a Glycan-Directed Antibody-Drug Conjugate. Cancer Res 2019; 79:1810-1821. [PMID: 30808675 PMCID: PMC6467748 DOI: 10.1158/0008-5472.can-18-3119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/01/2019] [Accepted: 02/21/2019] [Indexed: 12/23/2022]
Abstract
The specific targeting of differentially expressed glycans in malignant cells has emerged as an attractive anticancer strategy. One such target is the oncodevelopmental antigen polysialic acid (polySia), a polymer of α2,8-linked sialic acid residues that is largely absent during postnatal development but is re-expressed during progression of several malignant human tumors, including small-cell and non-small cell lung carcinomas, glioma, neuroblastoma, and pancreatic carcinoma. In these cancers, expression of polySia correlates with tumor progression and poor prognosis and appears to modulate cancer cell adhesion, invasiveness, and metastasis. To evaluate the potential of PolySia as a target for anticancer therapy, we developed a chimeric human polySia-specific mAb that retained low nanomolar (nmol/L) target affinity and exhibited exquisite selectivity for polySia structures. The engineered chimeric mAb recognized several polySia-positive tumor cell lines in vitro and induced rapid endocytosis of polySia antigens. To determine whether this internalization could be exploited for delivery of conjugated cytotoxic drugs, we generated an antibody-drug conjugate (ADC) by covalently linking the chimeric human mAb to the tubulin-binding maytansinoid DM1 using a bioorthogonal chemical reaction scheme. The resulting polySia-directed ADC demonstrated potent target-dependent cytotoxicity against polySia-positive tumor cells in vitro. Collectively, these results establish polySia as a valid cell-surface, cancer-specific target for glycan-directed ADC and contribute to a growing body of evidence that the tumor glycocalyx is a promising target for synthetic immunotherapies. SIGNIFICANCE: These findings describe a glycan-specific antibody-drug conjugate that establishes polySia as a viable cell surface target within the tumor glycocalyx.
Collapse
Affiliation(s)
- Emily C Cox
- Biological and Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York
| | - Dana N Thornlow
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Michaela A Jones
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Jordan L Fuller
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | | | - Matthew J Paszek
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Christopher A Alabi
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Matthew P DeLisa
- Biological and Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York.
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| |
Collapse
|
7
|
Pazynina GV, Tsygankova SV, Ryzhov IM, Paramonov AS, Nicolai, Bovin V. Synthesis of H (type 4) trisaccharide, key structural fragment of globo-H and fucosyl-GM1 cancer-associated antigens. MENDELEEV COMMUNICATIONS 2018. [DOI: 10.1016/j.mencom.2018.07.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
8
|
Dotz V, Wuhrer M. Histo-blood group glycans in the context of personalized medicine. Biochim Biophys Acta Gen Subj 2016; 1860:1596-607. [PMID: 26748235 PMCID: PMC7117023 DOI: 10.1016/j.bbagen.2015.12.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND A subset of histo-blood group antigens including ABO and Lewis are oligosaccharide structures which may be conjugated to lipids or proteins. They are known to be important recognition motifs not only in the context of blood transfusions, but also in infection and cancer development. SCOPE OF REVIEW Current knowledge on the molecular background and the implication of histo-blood group glycans in the prevention and therapy of infectious and non-communicable diseases, such as cancer and cardiovascular disease, is presented. MAJOR CONCLUSIONS Glycan-based histo-blood groups are associated with intestinal microbiota composition, the risk of various diseases as well as therapeutic success of, e.g., vaccination. Their potential as prebiotic or anti-microbial agents, as disease biomarkers and vaccine targets should be further investigated in future studies. For this, recent and future technological advancements will be of particular importance, especially with regard to the unambiguous structural characterization of the glycan portion in combination with information on the protein and lipid carriers of histo-blood group-active glycans in large cohorts. GENERAL SIGNIFICANCE Histo-blood group glycans have a unique linking position in the complex network of genes, oncodevelopmental biological processes, and disease mechanisms. Thus, they are highly promising targets for novel approaches in the field of personalized medicine. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.
Collapse
Affiliation(s)
- Viktoria Dotz
- Division of Bioanalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands; Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Manfred Wuhrer
- Division of Bioanalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands; Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
Sterner E, Flanagan N, Gildersleeve JC. Perspectives on Anti-Glycan Antibodies Gleaned from Development of a Community Resource Database. ACS Chem Biol 2016; 11:1773-83. [PMID: 27220698 PMCID: PMC4949583 DOI: 10.1021/acschembio.6b00244] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
![]()
Antibodies are used
extensively for a wide range of basic research
and clinical applications. While an abundant and diverse collection
of antibodies to protein antigens have been developed, good monoclonal
antibodies to carbohydrates are much less common. Moreover, it can
be difficult to determine if a particular antibody has the appropriate
specificity, which antibody is best suited for a given application,
and where to obtain that antibody. Herein, we provide an overview
of the current state of the field, discuss challenges for selecting
and using antiglycan antibodies, and summarize deficiencies in the
existing repertoire of antiglycan antibodies. This perspective was
enabled by collecting information from publications, databases, and
commercial entities and assembling it into a single database, referred
to as the Database of Anti-Glycan Reagents (DAGR). DAGR is a publicly
available, comprehensive resource for anticarbohydrate antibodies,
their applications, availability, and quality.
Collapse
Affiliation(s)
- Eric Sterner
- Chemical Biology Laboratory,
Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Natalie Flanagan
- Chemical Biology Laboratory,
Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Jeffrey C. Gildersleeve
- Chemical Biology Laboratory,
Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
10
|
Labrada M, Pablos I, Prete F, Hevia G, Clavell M, Benvenuti F, Fernández LE. Induction of leukocyte infiltration at metastatic site mediates the protective effect of NGcGM3-based vaccine. Hum Vaccin Immunother 2015; 10:2312-20. [PMID: 25424937 DOI: 10.4161/hv.29161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
While the NGcGM3/VSSP vaccine, a preparation consisting in very small sized proteoliposomes (VSSP) obtained by the incorporation of the NGcGM3 ganglioside into the outer membrane protein (OMP) complex of Neisseria meningitides, is currently studied in late stage clinical trials in breast cancer and melanoma patients, mechanisms involved in the vaccine's antitumor effect are insufficiently understood. Here we have addressed the role of adaptive and innate immune cells in mediating the protective effect of the vaccine. To this aim we selected the 3LL-D122 Lewis lung spontaneous metastasis model. Unexpectedly, inoculation of the vaccine in tumor bearing C57BL/6 mice, either by subcutaneous (sc) or intraperitoneal (ip) routes, induced similar anti-metastatic effect. Regardless the T-independent nature of NGcGM3 ganglioside as antigen, the antimetastatic effect of NGcGM3/VSSP is dependent on CD4(+) T cells. In a further step we found that the vaccine was able to promote the increase, maturation, and cytokine secretion of conventional DCs and the maturation of Bone Marrow-derived plasmacytoid DCs. In line with this result the in vivo IFNα serum level in ip vaccinated mice increased as soon as 2h after treatment. On the other hand the infiltration of NK1.1(+)CD3(-) and NK1.1(+)CD3(+) cells in lungs of vaccinated mice was significantly increased, compared with the presence of these cells in control animal lungs. In the same way NGcGM3/VSSP mobilized acquired immunity effector cells into the lungs of vaccinated tumor bearing mice. Finally and not less noteworthy, leukocyte infiltration in lungs of tumor bearing mice correlates with vaccine induced inhibition of lung metastization.
Collapse
Affiliation(s)
- Mayrel Labrada
- a Center of Molecular Immunology (CIM); Immunobiology Division; Atabey; Havana Cuba
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
Receptors expressed on the host cell surface adhere viruses to target cells and serve as determinants of viral tropism. Several viruses bind cell surface glycans to facilitate entry, but the contribution of specific glycan moieties to viral disease is incompletely understood. Reovirus provides a tractable experimental model for studies of viral neuropathogenesis. In newborn mice, serotype 1 (T1) reovirus causes hydrocephalus, whereas serotype 3 (T3) reovirus causes encephalitis. T1 and T3 reoviruses engage distinct glycans, suggesting that glycan-binding capacity contributes to these differences in pathogenesis. Using structure-guided mutagenesis, we engineered a mutant T1 reovirus incapable of binding the T1 reovirus-specific glycan receptor, GM2. The mutant virus induced substantially less hydrocephalus than wild-type virus, an effect phenocopied by wild-type virus infection of GM2-deficient mice. In comparison to wild-type virus, yields of mutant virus were diminished in cultured ependymal cells, the cell type that lines the brain ventricles. These findings suggest that GM2 engagement targets reovirus to ependymal cells in mice and illuminate the function of glycan engagement in reovirus serotype-dependent disease. Receptor utilization strongly influences viral disease, often dictating host range and target cell selection. Different reovirus serotypes bind to different glycans, but a precise function for these molecules in pathogenesis is unknown. We used type 1 (T1) reovirus deficient in binding the GM2 glycan and mice lacking GM2 to pinpoint a role for glycan engagement in hydrocephalus caused by T1 reovirus. This work indicates that engagement of a specific glycan can lead to infection of specific cells in the host and consequent disease at that site. Since reovirus is being developed as a vaccine vector and oncolytic agent, understanding reovirus-glycan interactions may allow manipulation of reovirus glycan-binding properties for therapeutic applications.
Collapse
|
12
|
Horwacik I, Rokita H. Targeting of tumor-associated gangliosides with antibodies affects signaling pathways and leads to cell death including apoptosis. Apoptosis 2015; 20:679-88. [DOI: 10.1007/s10495-015-1103-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
13
|
Colley KJ, Kitajima K, Sato C. Polysialic acid: biosynthesis, novel functions and applications. Crit Rev Biochem Mol Biol 2014; 49:498-532. [PMID: 25373518 DOI: 10.3109/10409238.2014.976606] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As an anti-adhesive, a reservoir for key biological molecules, and a modulator of signaling, polysialic acid (polySia) is critical for nervous system development and maintenance, promotes cancer metastasis, tissue regeneration and repair, and is implicated in psychiatric diseases. In this review, we focus on the biosynthesis and functions of mammalian polySia, and the use of polySia in therapeutic applications. PolySia modifies a small subset of mammalian glycoproteins, with the neural cell adhesion molecule, NCAM, serving as its major carrier. Studies show that mammalian polysialyltransferases employ a unique recognition mechanism to limit the addition of polySia to a select group of proteins. PolySia has long been considered an anti-adhesive molecule, and its impact on cell adhesion and signaling attributed directly to this property. However, recent studies have shown that polySia specifically binds neurotrophins, growth factors, and neurotransmitters and that this binding depends on chain length. This work highlights the importance of considering polySia quality and quantity, and not simply its presence or absence, as its various roles are explored. The capsular polySia of neuroinvasive bacteria allows these organisms to evade the host immune response. While this "stealth" characteristic has made meningitis vaccine development difficult, it has also made polySia a worthy replacement for polyetheylene glycol in the generation of therapeutic proteins with low immunogenicity and improved circulating half-lives. Bacterial polysialyltransferases are more promiscuous than the protein-specific mammalian enzymes, and new studies suggest that these enzymes have tremendous therapeutic potential, especially for strategies aimed at neural regeneration and tissue repair.
Collapse
Affiliation(s)
- Karen J Colley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, IL , USA and
| | | | | |
Collapse
|
14
|
Aiming at the sweet side of cancer: Aberrant glycosylation as possible target for personalized-medicine. Cancer Lett 2014; 352:102-12. [DOI: 10.1016/j.canlet.2013.10.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 10/09/2013] [Accepted: 10/09/2013] [Indexed: 01/26/2023]
|
15
|
Büll C, den Brok MH, Adema GJ. Sweet escape: sialic acids in tumor immune evasion. Biochim Biophys Acta Rev Cancer 2014; 1846:238-46. [PMID: 25026312 DOI: 10.1016/j.bbcan.2014.07.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/24/2022]
Abstract
Sialic acids represent a family of sugar molecules derived from neuraminic acid that frequently terminate glycan chains and contribute to many biological processes. Already five decades ago, aberrantly high expression of sialic acids has been proposed to protect cancer cells from recognition and eradication by the immune system. Today, increased understanding at the molecular level demonstrates the broad immunomodulatory capacity of tumor-derived sialic acids that is, at least in part, mediated through interactions with immunoinhibitory Siglec receptors. Here we will review current studies from a sialic acid sugar perspective showing that tumor-derived sialic acids disable major killing mechanisms of effector immune cells, trigger production of immune suppressive cytokines and dampen activation of antigen-presenting cells and subsequent induction of anti-tumor immune responses. Furthermore, strategies to modulate sialic acid expression in cancer cells to improve cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Christian Büll
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Martijn H den Brok
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Gosse J Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
16
|
|
17
|
Pio R, Corrales L, Lambris JD. The role of complement in tumor growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:229-62. [PMID: 24272362 DOI: 10.1007/978-1-4614-5915-6_11] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Complement is a central part of the immune system that has developed as a first defense against non-self cells. Neoplastic transformation is accompanied by an increased capacity of the malignant cells to activate complement. In fact, clinical data demonstrate complement activation in cancer patients. On the basis of the use of protective mechanisms by malignant cells, complement activation has traditionally been considered part of the body's immunosurveillance against cancer. Inhibitory mechanisms of complement activation allow cancer cells to escape from complement-mediated elimination and hamper the clinical efficacy of monoclonal antibody-based cancer immunotherapies. To overcome this limitation, many strategies have been developed with the goal of improving complement-mediated effector mechanisms. However, significant work in recent years has identified new and surprising roles for complement activation within the tumor microenvironment. Recent reports suggest that complement elements can promote tumor growth in the context of chronic inflammation. This chapter reviews the data describing the role of complement activation in cancer immunity, which offers insights that may aid the development of more effective therapeutic approaches to control cancer.
Collapse
Affiliation(s)
- Ruben Pio
- Oncology Division (CIMA), and Department of Biochemistry and Genetics (School of Science), University of Navarra, Pamplona, Spain,
| | | | | |
Collapse
|
18
|
Wu X, Ragupathi G, Panageas K, Hong F, Livingston PO. Accelerated tumor growth mediated by sublytic levels of antibody-induced complement activation is associated with activation of the PI3K/AKT survival pathway. Clin Cancer Res 2013; 19:4728-39. [PMID: 23833306 DOI: 10.1158/1078-0432.ccr-13-0088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE We addressed the possibility that low levels of tumor cell-bound antibodies targeting gangliosides might accelerate tumor growth. EXPERIMENTAL DESIGN To test this hypothesis, we treated mice with a range of monoclonal antibody (mAb) doses against GM2, GD2, GD3, and CD20 after challenge with tumors expressing these antigens and tested the activity of the same mAbs in vitro. We also explored the mechanisms behind the complement-mediated tumor growth acceleration that we observed and an approach to overcome it. RESULTS Serologically detectable levels of IgM-mAb against GM2 are able to delay or prevent tumor growth of high GM2 expressing cell lines both in vitro and in a SCID mouse model, whereas very low levels of this mAb resulted in slight but consistent acceleration of tumor growth in both settings. Surprisingly, this is not restricted to IgM mAb targeting GM2 but consistent against an IgG mAb targeting GD3 as well. These findings were mirrored by in vitro studies with antibodies against these antigens as well as GD2 and CD20 (with Rituxan), and shown to be complement-dependent in all cases. Complement-mediated accelerated growth of cultured tumor cell lines initiated by low mAb levels was associated with activation of the phosphoinositide 3-kinase (PI3K)/AKT survival pathway and significantly elevated levels of both p-AKT and p-PRAS40. This complement-mediated PI3K activation and accelerated tumor growth in vitro and in vivo are eliminated by PI3K inhibitors NVP-BEZ235 and Wortmannin. These PI3K inhibitors also significantly increased efficacy of high doses of these four mAbs. CONCLUSION Our findings suggest that manipulation of the PI3K/AKT pathway and its signaling network can significantly increase the potency of passively administered mAbs and vaccine-induced antibodies targeting a variety of tumor cell surface antigens.
Collapse
Affiliation(s)
- Xiaohong Wu
- Laboratory of Tumor Vaccinology, Melanoma Sarcoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | | |
Collapse
|
19
|
Pietanza MC, Rudin CM. Novel therapeutic approaches for small cell lung cancer: the future has arrived. Curr Probl Cancer 2012; 36:156-73. [PMID: 22495056 DOI: 10.1016/j.currproblcancer.2012.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Recent advances in developing synthetic carbohydrate-based vaccines for cancer immunotherapies. Future Med Chem 2012; 4:545-84. [DOI: 10.4155/fmc.11.193] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cancer cells can often be distinguished from healthy cells by the expression of unique carbohydrate sequences decorating the cell surface as a result of aberrant glycosyltransferase activity occurring within the cell; these unusual carbohydrates can be used as valuable immunological targets in modern vaccine designs to raise carbohydrate-specific antibodies. Many tumor antigens (e.g., GM2, Ley, globo H, sialyl Tn and TF) have been identified to date in a variety of cancers. Unfortunately, carbohydrates alone evoke poor immunogenicity, owing to their lack of ability in inducing T-cell-dependent immune responses. In order to enhance their immunogenicity and promote long-lasting immune responses, carbohydrates are often chemically modified to link to an immunogenic protein or peptide fragment for eliciting T-cell-dependent responses. This review will present a summary of efforts and advancements made to date on creating carbohydrate-based anticancer vaccines, and will include novel approaches to overcoming the poor immunogenicity of carbohydrate-based vaccines.
Collapse
|
21
|
Heimburg-Molinaro J, Lum M, Vijay G, Jain M, Almogren A, Rittenhouse-Olson K. Cancer vaccines and carbohydrate epitopes. Vaccine 2011; 29:8802-26. [PMID: 21964054 PMCID: PMC3208265 DOI: 10.1016/j.vaccine.2011.09.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 08/18/2011] [Accepted: 09/06/2011] [Indexed: 12/17/2022]
Abstract
Tumor-associated carbohydrate antigens (TACA) result from the aberrant glycosylation that is seen with transformation to a tumor cell. The carbohydrate antigens that have been found to be tumor-associated include the mucin related Tn, Sialyl Tn, and Thomsen-Friedenreich antigens, the blood group Lewis related Lewis(Y), Sialyl Lewis(X) and Sialyl Lewis(A), and Lewis(X) (also known as stage-specific embryonic antigen-1, SSEA-1), the glycosphingolipids Globo H and stage-specific embryonic antigen-3 (SSEA-3), the sialic acid containing glycosphingolipids, the gangliosides GD2, GD3, GM2, fucosyl GM1, and Neu5GcGM3, and polysialic acid. Recent developments have furthered our understanding of the T-independent type II response that is seen in response to carbohydrate antigens. The selection of a vaccine target antigen is based on not only the presence of the antigen in a variety of tumor tissues but also on the role this antigen plays in tumor growth and metastasis. These roles for TACAs are being elucidated. Newly acquired knowledge in understanding the T-independent immune response and in understanding the key roles that carbohydrates play in metastasis are being applied in attempts to develop an effective vaccine response to TACAs. The role of each of the above mentioned carbohydrate antigens in cancer growth and metastasis and vaccine attempts using these antigens will be described.
Collapse
Affiliation(s)
| | - Michelle Lum
- Department of Cellular and Molecular Biology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Geraldine Vijay
- University of Texas, MD Anderson Cancer Center, Houston, Texas 77030
| | - Miten Jain
- Department of Biomolecular Engineering, University of California Santa Cruz, CA 95064
| | - Adel Almogren
- Department Of Pathology, College of Medicine, King Saud University, Riyadh, 11461 Saudi Arabia
| | - Kate Rittenhouse-Olson
- Department Of Pathology, College of Medicine, King Saud University, Riyadh, 11461 Saudi Arabia
- Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, Buffalo, NY 14214
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, NY 14214
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| |
Collapse
|
22
|
Krug LM, Ragupathi G, Hood C, George C, Hong F, Shen R, Abrey L, Jennings HJ, Kris MG, Livingston PO. Immunization with N-propionyl polysialic acid-KLH conjugate in patients with small cell lung cancer is safe and induces IgM antibodies reactive with SCLC cells and bactericidal against group B meningococci. Cancer Immunol Immunother 2011; 61:9-18. [PMID: 21811785 DOI: 10.1007/s00262-011-1083-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/19/2011] [Indexed: 11/24/2022]
Abstract
PURPOSE Polysialic acid (polySA) is a polymer side chain bound to the neural cell adhesion molecule that is extensively expressed on the surface of small cell lung cancer (SCLC) cells. In our previous study, a robust antibody response was noted in patients with SCLC after vaccination with 30 μg of keyhole limpet hemocyanin (KLH)-conjugated N-propionylated (NP-) polySA, but peripheral neuropathy and ataxia were detected in several vaccinated patients. The objectives of the current trial were to establish the lowest optimal dose and to confirm the safety of the induction of antibodies against polySA with the NP-polySA vaccine. EXPERIMENTAL DESIGN Patients with SCLC who completed initial treatment and had no evidence of disease progression were injected with either 10 or 3 μg of NP-polySA conjugated to KLH and mixed with 100 μg of immunologic adjuvant (QS-21) at weeks 1, 2, 3, 4, 8, and 16. RESULTS Nine patients were enrolled at each of the two dose levels. Prior to vaccination, one patient in each group had low-titer antibodies against polysialic acid. All patients at the 10 μg vaccine dose level responded to vaccination with IgM antibody titers against polysialic acid (median titer 1/1,280 by ELISA), and all but one patient made IgM and IgG antibodies against the artificial vaccine immunogen, NP-polysialic acid (median titer 1/10,240). The antibody responses at the 3 μg vaccine dose level were lower; six of nine patients developed antibodies against polysialic acid (median titer 1/160). Post-vaccination sera from 6/9 and 3/9 patients in the 10 and 3 μg groups reacted strongly with human SCLC cells by fluorescent-activated cell sorting (FACS). Sera from all patients in the 10 μg dose group also had bactericidal activity against group B meningococci with rabbit complement. Self-limited grade 3 ataxia of unclear etiology was seen in 1 of 18 patients. CONCLUSIONS Vaccination with NP-polySA-KLH resulted in consistent high-titer antibody responses, with the 10 μg dose significantly more immunogenic than the 3 μg dose. This study establishes the lowest optimally immunogenic dose of NP-polysialic acid in this NP-polysialic acid-KLH conjugate vaccine to be at least 10 μg, and it establishes the vaccine's safety. We plan to incorporate NP-polySA into a polyvalent vaccine against SCLC with four glycolipid antigens also widely expressed in SCLC-GD2, GD3, fucosylated GM1, and globo H.
Collapse
Affiliation(s)
- Lee M Krug
- Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, 1275 York Ave., New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Li J, Dai G, Cheng YB, Qi X, Geng MY. Polysialylation promotes neural cell adhesion molecule-mediated cell migration in a fibroblast growth factor receptor-dependent manner, but independent of adhesion capability. Glycobiology 2011; 21:1010-8. [DOI: 10.1093/glycob/cwr020] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
24
|
Polysialic Acid Bioengineering of Cancer and Neuronal Cells by N-Acyl Sialic Acid Precursor Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 705:679-88. [DOI: 10.1007/978-1-4419-7877-6_36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
25
|
Simmons O, Magee M, Nemunaitis J. Current vaccine updates for lung cancer. Expert Rev Vaccines 2010; 9:323-35. [PMID: 20218860 DOI: 10.1586/erv.10.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Current treatments for lung cancer are far from optimal. Several immunotherapeutic strategies involving vaccines incorporating different tumor-associated antigens to induce immune responses against tumors are being tested in clinical trials internationally. Although small, benefits have indeed been observed from the early studies of these vaccines, and the future is looking brighter for lung cancer patients as a handful of these immunotherapies reach Phase III trials. In addition, optimizing the induced immune response by these vaccines has become a priority, and a number of techniques are being considered, including addition of adjuvants and combining vaccines, which affect synergy based on their mechanism of action. This review is an update on the current vaccines in production, the benefits observed from their most recent studies, and the upcoming plans for improvements in these immunotherapies.
Collapse
|
26
|
Labrada M, Clavell M, Bebelagua Y, León JD, Alonso DF, Gabri MR, Veloso RC, Vérez V, Fernández LE. Direct validation of NGcGM3 ganglioside as a new target for cancer immunotherapy. Expert Opin Biol Ther 2010; 10:153-62. [PMID: 20088712 DOI: 10.1517/14712590903443084] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE The target concept means not only an aberrant expression of a particular molecule in tumour tissues but also evidence of a clear therapeutic advantage, as a consequence of immune-intervention, in an antigen-positive relevant tumour model. Since we reported the presence of NGcGM3 ganglioside in human breast tumours years ago and though Phase I clinical trials of a ganglioside containing vaccine have been conducted, a definitive direct validation of this peculiar molecule as target for cancer immunotherapy has remained unperformed. METHODS Two animal models were used: leghorn chickens and C57BL/6 mice. The murine 3LL-D122 cell line, the derived subcutaneous tumours and metastatic lung lesions were processed for gangliosides identification. Active immunotherapy experiments in the 3LL-D122 spontaneous lung metastasis model were performed with NGcGM3/VSSP vaccine prepared by conjugation of NGcGM3 with the outer membrane proteins of Neisseria meningitides. RESULTS The 3LL-D122 Lewis lung carcinoma results were consistent with an increased expression of NGcGM3 from primary tumours to metastatic lesions, as observed in human breast cancer samples. Both vaccines, prepared with synthetic or natural-source-derived ganglioside, showed similar anti-tumour and immunogenicity profiles. Finally, a clear involvement of NK1.1(+) cells and CD8(+) T cells in the anti-metastatic effect elicited by the vaccine was manifested. CONCLUSIONS While 'proof of concept' Phase II and III clinical trials with the NGcGM3/VSSP vaccine in cancer patients are currently ongoing these results reasonably sustain the validation of this peculiar ganglioside as a novel target for cancer immunotherapy.
Collapse
Affiliation(s)
- Mayrel Labrada
- Center of Molecular Immunology, Vaccine Department, 216 St and 15th Ave., Atabey, Playa, P. O. Box 16040, Havana, 11600, Cuba.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, Mellman I, Prindiville SA, Viner JL, Weiner LM, Matrisian LM. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res 2009; 15:5323-37. [PMID: 19723653 DOI: 10.1158/1078-0432.ccr-09-0737] [Citation(s) in RCA: 1026] [Impact Index Per Article: 64.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The purpose of the National Cancer Institute pilot project to prioritize cancer antigens was to develop a well-vetted, priority-ranked list of cancer vaccine target antigens based on predefined and preweighted objective criteria. An additional aim was for the National Cancer Institute to test a new approach for prioritizing translational research opportunities based on an analytic hierarchy process for dealing with complex decisions. Antigen prioritization involved developing a list of "ideal" cancer antigen criteria/characteristics, assigning relative weights to those criteria using pairwise comparisons, selecting 75 representative antigens for comparison and ranking, assembling information on the predefined criteria for the selected antigens, and ranking the antigens based on the predefined, preweighted criteria. Using the pairwise approach, the result of criteria weighting, in descending order, was as follows: (a) therapeutic function, (b) immunogenicity, (c) role of the antigen in oncogenicity, (d) specificity, (e) expression level and percent of antigen-positive cells, (f) stem cell expression, (g) number of patients with antigen-positive cancers, (h) number of antigenic epitopes, and (i) cellular location of antigen expression. None of the 75 antigens had all of the characteristics of the ideal cancer antigen. However, 46 were immunogenic in clinical trials and 20 of them had suggestive clinical efficacy in the "therapeutic function" category. These findings reflect the current status of the cancer vaccine field, highlight the possibility that additional organized efforts and funding would accelerate the development of therapeutically effective cancer vaccines, and accentuate the need for prioritization.
Collapse
Affiliation(s)
- Martin A Cheever
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yin J, Miyazaki K, Shaner RL, Merrill AH, Kannagi R. Altered sphingolipid metabolism induced by tumor hypoxia - new vistas in glycolipid tumor markers. FEBS Lett 2009; 584:1872-8. [PMID: 19913543 DOI: 10.1016/j.febslet.2009.11.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/07/2009] [Accepted: 11/09/2009] [Indexed: 02/06/2023]
Abstract
Uncontrolled growth of malignant cells produces hypoxic regions in locally advanced tumors. Recently we showed that tumor hypoxia-induced transcription of multiple genes involved in glycan synthesis, leading to expression of useful glycolipid tumor markers, such as gangliosides having N-glycolyl sialic acid. Our subsequent studies indicated that the ceramide portion of glycolipids, as well as their glycan moiety, was also significantly affected by hypoxia. Tumor hypoxia-induced marked accumulation of sphinganine (dihydrosphingosine) long-chain base, and significant reduction of unsaturated very long-chain fatty acids in the ceramide moiety. Mass-spectrometry, which yields information on both glycan- and ceramide moieties, is expected to be clinically useful in detecting such distinct molecular species of cancer-associated glycolipids having combined alteration in both glycan- and ceramide moieties.
Collapse
Affiliation(s)
- Jun Yin
- Department of Molecular Pathology, Aichi Cancer Center, Nagoya 464-8681, Japan
| | | | | | | | | |
Collapse
|
29
|
The Novel Chimeric Anti-NCAM (Neural Cell Adhesion Molecule) Antibody ch.MK1 Displays Antitumor Activity in SCID Mice but Does not Activate Complement-Dependent Cytolysis (CDC). J Immunother 2009; 32:442-51. [DOI: 10.1097/cji.0b013e31819f8b69] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
30
|
Schietinger A, Philip M, Schreiber H. Specificity in cancer immunotherapy. Semin Immunol 2008; 20:276-85. [PMID: 18684640 DOI: 10.1016/j.smim.2008.07.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Revised: 06/29/2008] [Accepted: 07/01/2008] [Indexed: 11/29/2022]
Abstract
From the earliest days in the field of tumor immunology three questions have been asked: do cancer cells express tumor-specific antigens, does the immune system recognize these antigens and if so, what is their biochemical nature? We now know that truly tumor-specific antigens exist, that they are caused by somatic mutations, and that these antigens can induce both humoral and cell-mediated immune responses. Because tumor-specific antigens are exclusively expressed by the cancer cell and are often crucial for tumorigenicity, they are ideal targets for anti-cancer immunotherapy. Nevertheless, the antigens that are targeted today by anti-tumor immunotherapy are not tumor-specific antigens, but antigens that are normal molecules also expressed by normal tissues (so-called "tumor-associated" antigens). If tumor-specific antigens exist and are ideal targets for immunotherapy, why are they not being targeted? In this review, we summarize current knowledge of tumor-specific antigens: their identification, immunological relevance and clinical use. We discuss novel tumor-specific epitopes and propose new approaches that could improve the success of cancer immunotherapy, especially for the treatment of solid tumors.
Collapse
Affiliation(s)
- Andrea Schietinger
- Department of Pathology and Committee on Immunology, The University of Chicago, 5841 South Maryland Avenue MC 3008, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
31
|
Furukawa K, Hamamura K, Nakashima H, Furukawa K. Molecules in the signaling pathway activated by gangliosides can be targets of therapeutics for malignant melanomas. Proteomics 2008; 8:3312-6. [DOI: 10.1002/pmic.200800228] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Kannagi R, Yin J, Miyazaki K, Izawa M. Current relevance of incomplete synthesis and neo-synthesis for cancer-associated alteration of carbohydrate determinants--Hakomori's concepts revisited. Biochim Biophys Acta Gen Subj 2007; 1780:525-31. [PMID: 17980710 DOI: 10.1016/j.bbagen.2007.10.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2007] [Accepted: 10/12/2007] [Indexed: 12/12/2022]
Abstract
Incomplete synthesis and neo-synthesis are two major concepts for cancer-associated alterations of cell surface carbohydrate determinants, formulated by Hakomori and collaborators almost 25 years ago. These concepts are still as relevant and useful as ever for cancer-associated alteration of carbohydrate determinants. Incomplete synthesis of carbohydrate determinants occurs through the epigenetic silencing of glycogenes through DNA methylation and/or histone modification in the early stage cancers. The natural selection of more malignant cancer cells occurs through acquisition of hypoxia resistance by constitutively activated hypoxia inducible factors (HIFs) in the advanced stages of cancers. HIFs induce transcription of several important glycogenes, and lead to neo-synthesis of carbohydrate determinants. For instance, expression of sialyl Lewis A/X is induced by epigenetic silencing of glycogenes in the early stages, and is further accelerated in the advanced stages by hypoxia-induced transcription of several glycogenes. Expression of GM2 ganglioside is induced in cancers by altered glycosyltransferase activities, and its N-glycolyl sialic acid content increases by hypoxia-induced transcription of a sialic acid transporter gene. N-glycolyl GM2 thus reflects two cancer-associated genetic abnormalities in a single determinant, and has high cancer specificity. Every carbohydrate determinant is synthesized through multiple steps, each of which is affected by cancer-associated genetic abnormality. Superiority of carbohydrate determinants as cancer-specific molecules over protein determinants is demonstrated in that a single carbohydrate determinant can reflect multiple cancer-associated genetic abnormalities.
Collapse
Affiliation(s)
- Reiji Kannagi
- Department of Molecular Pathology, Research Institute, Aichi Cancer Center, 1-1 Kanokoden, Chikusaku, Nagoya 464-8681, Japan.
| | | | | | | |
Collapse
|
33
|
Ellison BS, Zanin MKB, Boackle RJ. Complement susceptibility in glutamine deprived breast cancer cells. Cell Div 2007; 2:20. [PMID: 17623109 PMCID: PMC2031881 DOI: 10.1186/1747-1028-2-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 07/11/2007] [Indexed: 11/17/2022] Open
Abstract
Background Membrane complement regulatory proteins (mCRPs) inhibit complement-mediated killing of human cells by human complement, a property that confers protection from complement to malignant breast cancer cells and that thwarts some immunotherapies. Metabolic mechanisms may come into play in protecting cancer cells from the complement system subsequent to relatively low levels of complement deposition. Results In differentiating these mechanisms, two types of human breast cancer cell lines, MCF7 (adenocarcinoma) and Bcap37 (medullary carcinoma) were cell-cycle synchronized using glutamine-deprivation followed by restoration. These cells were examined for the expression of two mCRPs (CD59 and CD55), and for subsequent susceptibility to antibody-mediated complement-induced membrane damage. After glutamine restoration, MCF7 and Bcap37 cells were synchronized into the G2/M phase and an average increased expression of CD59 and CD55 occurred with a corresponding resistance to complement-mediated damage. Blocking CD59 inhibitory function with monoclonal antibody revealed that CD59 played a key role in protecting unsynchronized Bcap37 and MCF7 cancer cells from the complement membrane attack complex. Interestingly, glutamine-deprivation did not significantly affect the expression of proteins e.g., the surface level of CD59 or CD55, but did increase the susceptibility to complement-mediated killing. One possible explanation is that glutamine-deprivation may have slowed the turnover rate of mCRPs, preventing the cells from replacing pre-existing mCRPs, as they became neutralized by covalent C4b and C3b depositions. Conclusion Taken together the findings are consistent with the conclusion that future immunotherapies should aim to achieve a highly specific and profound activation and deposition of complement as well as to disrupt the synthesis and expression of CD59 and CD55 by the cancer cells.
Collapse
Affiliation(s)
- Bradley S Ellison
- Department of Orthopaedic Surgery, The Ohio State University, 410W 10th Ave, Columbus Ohio 43210, USA
| | - Mary KB Zanin
- Department of Biology, The Citadel, 171 Moultrie Street, Charleston, SC 29409, USA
| | - Robert J Boackle
- Department of Stomatology and Department of Immunology, Medical University of South Carolina, 171 Ashley Ave., Charleston, South Carolina 29425, USA
| |
Collapse
|
34
|
Tokuda N, Zhang Q, Yoshida S, Kusunoki S, Urano T, Furukawa K, Furukawa K. Genetic mechanisms for the synthesis of fucosyl GM1 in small cell lung cancer cell lines. Glycobiology 2006; 16:916-25. [PMID: 16880505 DOI: 10.1093/glycob/cwl022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fucosyl GM1 has been reported to be specifically expressed in small cell lung cancer (SCLC) cells. However, the genetic basis for the synthesis of fucosyl GM1 has not been investigated. We analyzed the glycosyltransferases responsible for the synthesis of fucosyl GM1 in SCLC cell lines. In four SCLC cell lines expressing fucosyl GM1, both FUT1 and FUT2 mRNAs were detected, indicating that either one or both of alpha1,2-fucosyltransferases may be involved in the expression of fucosyl GM1. However, three of these four lines contained function-loss mutations in the FUT2 coding region, suggesting that FUT1 is mainly involved in the alpha1,2-fucosylation of GM1. The expression levels of the GM1 synthase gene showed no correlation with those of fucosyl GM1, whereas the co-transfection of GM1 synthase cDNA with FUT1 or FUT2 into SK-LC-17 clearly enhanced the neo-expression of fucosyl GM1, indicating its essential role. In contrast, the co-transfection of GD3 synthase cDNA reduced the expression levels of fucosyl GM1 with FUT1 or FUT2. Consequently, FUT1 seems to mainly contribute to the expression of fucosyl GM1, although both FUT1 and FUT2 are capable of generating the antigen. These results should promote the functional analysis of fucosyl GM1 leading to the development of novel therapies for SCLC.
Collapse
Affiliation(s)
- Noriyo Tokuda
- Department of Biochemistry II, Nagoya University School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|