Abstract
Tumor cells are characterized by the expression of tumor-specific carbohydrate structures that differ from their normal counterparts. Carbohydrates on tumor cells have phenotypical as well as functional implications, impacting the tumor progression process, from malignant transformation to metastasis formation. Importantly, carbohydrates are structures that play a role in receptor–ligand interaction and elicit the activity of growth factor receptors, integrins, lectins, and other type 1 transmembrane proteins. They have been recognized as biomarkers for cancer diagnosis, and evidence demonstrating their relevance as targets for anticancer therapeutic strategies, including immunotherapy, continues to accumulate. Different approaches targeting carbohydrates include monoclonal antibodies (mAbs), antibody (Ab)–drug conjugates, vaccines, and adhesion antagonists. Development of bispecific antibodies and chimeric antigen receptor (CAR)-modified T cells against tumor-associated carbohydrate antigens (TACAs) as promising cancer immunotherapeutic agents is rapidly evolving. As reviewed here, there are several cancer-associated glycan features that can be leveraged to design rational drug or immune system targets, applying multiple TACA structural and functional features to be targeted as the standard treatment paradigm. Many of the underlying targets were defined by researchers at the Wistar Institute in Philadelphia, Pennsylvania, which provide basis for different immunotherapy approaches.
Keywords: monoclonal antibody, carbohydrate-targeted immunotherapy, cancer vaccines, lectins, cancer diagnostic, tumor-associated carbohydrate antigens (TACA)
Introduction
Alteration in glycosylation is a proven characteristic of cancer, and most known serological biomarkers currently used in the clinic are cancer-associated glycans. Biosynthesis of glycans is non-DNA template dependent and it is controlled by the expression of glycosyltransferases and glycosidases. Tissues of different origin and cancer cells express different carbohydrate structures due to expression of these enzymes. Aberrant glycosylation can include sialylation, fucosylation, O-glycan truncation or incomplete synthesis, and N- and O-linked glycan branching.(1) Both aberrant expression of carbohydrates and acquisition of aberrant glycosylation profiles accompany malignant transformation and progression.(2) Carbohydrates are the most diverse complex biomolecules that also play pivotal roles in many cellular physiological functions, including cell–cell interaction, cellular signaling through cell surface receptors, and immune recognition. Although tumor-specific glycans have been known for a long time (Table 1), only recently, these structures have been identified as potential targets to recruit the host immune system for cancer therapy or generate the immune response through vaccines.
Table 1.
Tumor-associated carbohydrate antigens | Structure |
---|---|
Tn | GalNAcSer/Thr |
sialyl Tn | Neu5Acα2-6GalNAcaSer/Thr |
T antigen | Galβ1-3GalNAcα1-Ser/Thr |
Globo-H | Fucα1-2Galβ1-3GalNAcβ1-3Galα1-4Galβ1-4Glc |
LeY | Fuc(α1 → 2)Gal(β1 → 4)[Fuc(α1 → 3)]GlcNAc |
SLea | Neu5Acα2,3Galβ1,3(Fucα1,4)GlcNAc |
SLeX | Neu5Acα2,3Galβ1,4 (Fucα1,3)GlcNAc |
GM3 | Neu5Acα2,3Galβ1,4Glcβ1Cer |
GD3 | Neu5Acα2,8Neu5Acα2,3Galβ1, 4Glcβ1Cer |
GD2 | GalNAcβ1,4(Neu5Acα2,8Neu5Acα2,3)Galβ1, 4Glcβ1Cer |
LeY, Lewis Y; SLea, sialyl Lewis A; SLeX, sialyl Lewis X; STn, sialyl Tn.
Common approaches for immunotherapy include monoclonal antibodies (mAbs), vaccines, carbohydrate-specific antibody (Ab)–drug conjugates, carbohydrate mimetics, antagonists targeting selectins, and Siglec receptors. For example, several lectin families play a role in inflammatory processes and cancer, including selectins, galectins, siglecs, and macrophage galactose-type lectin (MGL). Glycan–lectin interactions are critical for cancer progression, cell proliferation, extravasation, and invasion. Targeting the glycans and interference in their interactions with specific inhibitors is currently being explored in clinical trials as a promising therapy strategy (Table 2).
Table 2.
Modality | Target | Histology | Phase | Clinical Trial |
---|---|---|---|---|
Vaccines | ||||
Glycolipids/Glycoproteins | Theratope STn Thomsen-Friedenreich (TF) |
Breast Prostate |
3 1 |
NCT00003638 NCT00003819 |
Globo-H-GM2-sTn-TF-Tn | Ovarian | 1 | NCT01248273 | |
GD2, GD3, Globo H, Fucosyl GM1 and N-propionylated polysialic acid | Lung | 1 | NCT01349647 | |
Globo-H | Breast | 2 | NCT01516307 | |
GM2, GD2, GD3 | Sarcoma | 2 | NCT01141491 | |
GD3 | Melanoma | 2 1 |
NCT00679289, NCT03159117 |
|
GD3L, GD3L | Melanoma | 1 1 |
NCT00597272 NCT00911560 |
|
GM2 | Breast Melanoma |
3 3 |
NCT00003357 NCT00005052 |
|
Anti-idiotype | NeuGcGM3 ACA125 CEA GD2 |
NSCLC Pediatric tumors Ovarian, Colorectal Neuroblastoma |
3 1 2 2 1 |
NCT01460472 NCT01598454 NCT00058435 NCT00033748 NCT00003023 |
Peptide Carbohydrate Mimotope | LeY, GD2 | Breast Lung |
1 and 2 1/2 |
NCT02229084 NCT02264236 |
Monoclonal Antibodies | ||||
GD2 | Neuroblastoma Neuroblastoma/ Osteosarcoma |
1 1 and 2 2 2 2 3 1 and 2 |
NCT03033303 NCT01757626 NCT00089258 NCT03363373 NCT00002458 NCT01704716 NCT03860207 |
|
GD3 | Melanoma | 2 | NCT00679289 | |
LeY | Breast Ovarian |
2 2 2 |
NCT01370239 NCT01370239 NCT00617773 |
|
SLea | Pancreas Gastrointestinal cancers |
1 2 |
NCT02672917 NCT03801915 |
|
MUC1 | Ovarian, Solid tumors |
2 1 |
NCT01899599 NCT01222624 |
|
Radiolabeled mAb | GD2 | Central Nervous System, Lung, Melanoma, Neuroblastoma, Sarcoma | 2 | NCT00445965 |
Antibody drug conjugate | LeY | NSCLC Prostate Ovarian Breast |
2 2 2 2 |
NCT00051571 NCT00031187 NCT00051584 NCT00028483 |
Lectin antagonists | ||||
Selectins | Acute Myeloid Leukemia | 3 | NCT03616470 | |
Galectins | Melanoma, NSCLC, H&N, Colon cancer, Solid tumors Melanoma |
1 2 1 1 and 2 1 |
NCT02575404, NCT00110721, NCT00054977, NCT01723813, NCT02117362 |
|
Siglec-15 | H&N, NSCLC Ovarian TNBC |
1/2 | NCT03665285 | |
CAR T cells | ||||
LeY | Solid tumors Myeloid malignancies |
1 1 and 2 |
NCT03851146 NCT02958384 |
|
CEA | Pancreatic | 2 and 3 |
NCT04037241 NCT03818165 |
|
GD2 | Glioma Neuroblastoma, Osteosarcoma B cell lymphoma |
1 1 1 1 and 2 1 and 2 1 1 1 and 2 |
NCT04099797 NCT04196413 NCT04099797 NCT04637503 NCT03373097 NCT01953900 NCT02107963 NCT04429438 |
|
Bispecific Antibodies | GD2 | Neuroblastoma | 1 and 2 | NCT02173093 |
CAR, chimeric antigen receptor; CEA, carcinoembryonic antigen; H&N, head and neck cancer; mAbs, monoclonal antibodies; NB, neuroblastoma; NSCLC, non small cell lung cancer; sTn, sialyl Tn; TNBC, triple negative breast cancer.
Research into tumor-associated carbohydrate antigens (TACAs) has entered an exciting phase because of the recent identification of their function and implications for clinical use. These discoveries open up the possibility of using TACAs and mAbs recognizing TACAs in vaccines and immunotherapeutic strategies against cancer. Pioneering studies focusing on cancer-associated cell surface glycans have been carried out using hybridoma technology and mAbs developed at the Wistar Institute of Anatomy and Biology starting in the late 1970s.(3–5) Future studies were performed in collaboration with the Department of Medical Biochemistry, University of Goteborg in Sweden; Department of Chemistry, University of Alberta, Edmonton, Canada; University of Pennsylvania, Philadelphia, Pennsylvania; and the National Institutes of Health (NIH), Bethesda, Maryland. These studies succeeded in identification of multiple carbohydrate tumor-associated antigens (Ags), including the first glycolipid tumor-associated antigen, sialyl Lewis A (SLea), known as CA19-9.(6,7) The SLea-specific antibody, named NS19-9, was developed at Wistar Institute by hybridoma technology using the human colorectal cancer cell line, SW1116, as an immunizing antigen.(4) The SLea antigen is widely expressed in gastrointestinal cancers and is one of the most studied serum biomarkers for diagnosis and monitoring of pancreatic and colorectal cancers. CA19-9 carbohydrate also plays a role in tumor extravasation through interaction with selectins, as discussed below. A recent study postulates that SLea promotes pancreatic cancer. This study demonstrated the possibility of CA19-9 as a therapeutic target for treatment of pancreatitis and pancreatic cancer.(8) Consequently, the CA19-9 antigen itself, its mimic, and the mAb are utilized for various immunotherapy approaches.
Examples of TACAs found by this group represent blood group-related carbohydrate series such as A, B, O, and Lewis (Le) system series, including sialyl-Lea (SLea), sialyl Lewis X (SLeX), and Lewis Y (LeY), which are widely expressed in epithelial tumors (Table 1).(9–20) SSEA-1 (CD15 or Lex) was also identified as a stage-specific embryonic antigen at the Wistar Institute using hybridoma technology.(21,22)
The ability to detect these aberrant glycans in situ in FFPE specimens using carbohydrate-specific mAbs has also been demonstrated by our group. These data indicate that the blood group ABO, H, Se, and Le genes are subjected to a tissue-dependent differential expression. The results of these studies laid the groundwork to evaluate blood group Ags and related glycolipids as pathological tumor markers and provide immunohistochemical evidence for a diverse repertoire of altered antigen expression in different cancers, which can be exploited for diagnosis and therapeutic intervention.(23–26)
Tumor tissues can also display gangliosides such as GD2, GD3, GM3, GM2, fucosyl GM1, and Globo-H that are sialylated glycosphingolipids found at elevated levels in tumors of neuroectodermal origin, including neuroblastomas (NBs) and melanomas. Specifically, a gradual increase in GD2, GD3, and 9-0-acetyl-GD3 ganglioside expression in subsequent stages of melanoma progression from normal melanocytes to metastatic disease, including the pivotal step of the early primary melanoma in the radial growth phase (RGP) to advanced vertical growth phase (VGP) melanoma, was characterized. The qualitative differences of gangliosides between the RPG and VGP suggest their role as prognostic indicators of risk for tumor recurrence and as a therapy target.(27–30) A phase I clinical trial has been conducted with murine mAb ME361, which recognizes GD2 and GD3 generated at the Wistar Institute.(31) The initial study, including clinical trials from this and other groups, built the foundation to further exploit the therapeutic and diagnostic potential for ganglioside-expressing tumors.
The assembly of cell surface complex carbohydrates requires the concerted action of a large number (>100) of glycosyltransferases, each of which catalyzes the transfer of a single sugar residue, usually from a sugar nucleotide, to specific hydroxyl groups on a suitable oligosaccharide acceptor. Glycosyltransferases such as sialyltransferases and fucosyltransferases involved in linking terminating residues on glycans are two of the most common glycosylation changes in carcinogenesis and progression. The increase in activity of these glycosyltransferases leads to overexpression of terminal TACA epitopes commonly found on transformed cells that include SLeX, SLea, sialyl Tn (STn), Globo H, LeY, and gangliosides.(32,33) α-2-L-Fucosyltransferase transfers L-fucose from GDP-L-fucose to the C-2 position of terminal nonreducing b-D-galactosyl residues, thus forming the H antigen from its type 1 or 2 chain precursor.
Our group characterized kinetic and structural parameters of both Secretor (Se) and H α-2-L-fucosyltransferases that are responsible for the synthesis of H (O-type) blood group and Lewis series Ags.(34,35) We published the amino acid sequence for α-2-L-fucosyltransferase and demonstrated that the enzyme-enhanced expression correlated with colon cancer progression.(36) The elevated level of the enzyme in adenomatous polyps may represent an early event associated with tumorigenesis in colon cancer. A nucleotide sequence analysis of the protein coding region of the complementary DNAs (cDNAs) derived from adenoma, and colon adenocarcinoma revealed 100% homology, suggesting that there is no tumor-associated allelic variant within the H α-2-L-fucosyltransferase cDNA.(37)
Glycosyltransferases represent prime targets for the design of glycosylation inhibitors with the potential to specifically alter the structures of cell surface carbohydrates. The study by our group on the mechanism of glycosyl transfer demonstrated that the reactive acceptor hydroxyl groups are involved in a critical hydrogen bond donor interaction with a basic group on the enzyme, which removes the developing proton during the glycosyl transfer reaction. The resulting deoxygenated acceptor analogs can no longer be substrates for the corresponding glycosyltransferases, which should act as competitive inhibitors. Alternatively, basic groups would be logical targets for irreversible covalent inactivation of the enzymes. Inhibitors of glycosylation can be invaluable tools in deciphering both the biosynthetic pathways for the assembly of active cell surface oligosaccharides, as well as tools for drug discovery.(38)
Defining the epitopes for antibodies and T cell receptors (TCRs) is of great importance for optimization of antigenic and immunogenic properties of effective vaccines and other immunotherapeutic approaches based on the Ab or TCR antigen recognition. To understand the basic principles of antibody-targeting TACAs and their binding specificity, structural studies, including biochemical methods, mass spectrometry, and proton nuclear magnetic resonance (NMR) spectroscopy, as well as conformations established by computer modeling, were undertaken by our group.(39–44)
While missing detailed crystallographic information, topographical features of antibody recognition and the conformational properties of a series of related tissue blood group (Lewis) carbohydrates were established. For example, using two-dimensional NMR in combination with hard-sphere energy calculation, it was established that the NS-19-9 antibody does not cross-react with Lea antigen and the presence of Neu5Ac residue can cause conformational alteration, which are crucial to the formation of the antigenic determinant.(39) Combining molecular modeling and experimental structural information may be possible to rationally modify Lewis antigen-binding antibodies by fine-tuning specificities and affinities to optimize their in vivo functionalities.(45,46) Importantly, these studies were conducted at the time when the methods of cocrystallizing antibody Fab fragments and carbohydrate antigens had not yet been used for antibody recognition and the conformational properties of blood group (Lewis) carbohydrates.(47,48)
The efficacy of the antibody can be further improved by increasing the specificity for the carbohydrate antigen that may be important for antibody-based approaches, including chimeric antigen receptor (CAR)-modified T cells, as even minor alterations in binding of the carbohydrate structure may have an impact on the bound conformations and affinity of the antigen important for recognition of the tumor antigen.
Recent efforts on carbohydrate-based cancer immunotherapies, including bispecific antibodies (BsAbs) and CAR T cells against TACAs, are rapidly evolving. This perspective discusses the role of carbohydrates for the current applications in oncology, including diagnostics and immunotherapy approaches. We also would like to acknowledge that many of today's modern cancer drugs may owe their conceptual basis to the pioneering work by Koprowski's team at the Wistar Institute decades ago, which can inform current and future developments in the field, including carbohydrate Ags as diagnostics, mAbs as drugs, vaccines, adhesion antagonists, and even BsAbs and CARs.
Carbohydrates as Diagnostics
Current clinically approved serological biomarkers for cancer diagnosis and biomarkers of disease recurrence in different cancers are glycosylated biomarkers. Most of the clinically relevant glycoprotein biomarkers in cancer patients include the prostate-specific antigen (PSA); carcinoembryonic antigen (CEA); ovarian carcinoma antigen CA125 also known as MUC16; breast cancer CA15-3, that is, aberrantly glycosylated MUC1; CA72-4 antigen in gastric cancer alpha fetoprotein (AFP) in liver cancer; and breast cancer antigen CA27-29.(49) Early studies from our group introduced mAbs for serological detection of TACAs.(50–55) The SLea antigen expressed in epithelial tumors is detected by the serological assay NS19-9 used for patients with an established diagnosis of pancreatic, colorectal, gastric, or biliary cancer and is used to monitor clinical response to therapy. It is the most clinically validated serum biomarker used for the management of pancreatic cancer patients to date.(56,57) One of the most important limitations of SLea as a tumor marker is that 5%–10% of the population lacks the ability to synthesize the SLea precursor due to failure of the Lewis α-4-L-fucosyltransferase expression and, as a result, cannot produce CA19-9, as noted in our report.(58)
Carbohydrate Targets and mAbs for Immunotherapy
Multiple approaches targeting carbohydrates have been investigated, and multiple clinical trials support the potential of targeting glycosylation in cancer immunotherapy. The comprehensive list of clinical trials applying different approaches targeting cancer-specific glycans has been listed in Table 2.(59,60) Considering the therapeutic approaches in which TACAs are targeted, their expression should be required as an eligibility criterion for patient stratification or selection to personalize patients' clinical outcomes.
Monoclonal Antibodies
Developing antibodies against TACAs for clinical use has been challenging and few tumor-targeting antibodies have reached clinical trials due to low affinity and toxicity. Nevertheless, several glycan-specific mAbs and their chimeric and/or humanized versions showed promise in in vitro and in vivo models and have been used for passive immunotherapy in clinical protocols. Several mAbs targeting glycolipids, such as GD2, GD3, GM2, LeY, or SLea, have demonstrated the ability to mediate potent antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CMC). The efficacy of mAb treatment was often enhanced with combination therapies or antibody-mediated delivery of cytotoxic payloads such as radioisotope and antibody–drug/toxin conjugates or cytokines such as granulocyte–macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-2).
As protein engineering technology has become more accessible, newer antibody constructs are being tested in clinical trials. Fully human antibodies were derived from lymphocytes of individuals immunized with the SLea-KLH vaccine demonstrated to be potent in CMC and ADCC.(61,62) SLea-targeting antibody MVT-5873 is currently in phase I clinical trial (NCT02672917) and have been reported to be well tolerated. A phase II trial was designed to determine the efficacy and safety of peri-operative Ab use in patients with gastrointestinal cancers (NCT03801915). Promising antibodies with high affinity for SLea (5B1 and 7E3) were also expressed as recombinant antibodies and were potent in CDC ADCC assays.(63) The most recent example demonstrates that reengineering the Fc portion of SLea-targeting antibodies generated in response to the SLea/KLH vaccine enhanced their affinity for activating human FcγRs and led to an enhanced therapeutic effect suggesting that an Fc-optimized variant could potentially be translated to the clinic.(64) Potentially natural class-switch variants can also show enhanced therapeutic potential. Spontaneously occurring mAb specific for SLea and LeY heavy chain variants, IgG1, IgG2b, and IgG2a, identified in our study had the same binding specificity and were active in ADCC; however, IgG2a gave the highest percentage of lysis and inhibited growth of human colorectal carcinoma, while IgGl and IgG2b were ineffective.(65–67) The results demonstrate the potential usefulness of the subclass switching antibody as a therapeutic agent.
Although GD2 and GD3-specific mAbs demonstrated in vivo and in vitro activity against melanoma and NB, the concerns with toxicity and low efficacy of mAbs prompted new advances in genetic engineering and development of newer generation chimeric and humanized anti-GD2 mAbs. The human–murine chimeric Ab ch14.18, subsequently renamed dinutuximab, was generated by combining the variable regions of the original murine IgG3 anti-GD2 mAb 14.18 and the constant regions of human IgG1.(68,69) Dinutuximab was the first antibody to be approved by the US FDA and the European Medicines Agency (EMA) for NB and other pediatric solid tumors and was established as the new standard of care for maintenance therapy in patients with high-risk NB. Phase 2 and 3 studies are underway for NB NCT03363373 NCT00002458 NCT01704716.
The LeY antigen is also a member of a family of blood group antigens expressed in human epithelial cancers, which makes it an attractive target for treatment with mAbs. Humanized or chimeric forms of LeY-specific mAbs have demonstrated signal of efficacy in preclinical models and have been tested in clinical trials.(70) The humanized mAb against the LeY antigen (Hu3S193) has been demonstrated to be safe in previous studies and has also been indicated as a potential intervention in solid tumors.(71–73) Hu3S193 is a humanized anti-LeY IgG1 mAb with strong complement and antibody-dependent cytotoxicity with clinical benefit shown in a phase II study (NCT00617773).
Other applications of TACA-specific mAbs include drugs delivered through antibody–drug conjugates (ADCs). LeY-specific BR96 mAbs conjugated to doxorubicin and docetaxel were tested in phase II trials for advanced non small cell lung cancer (NSCLC) and breast cancer.(74,75) Multiple ADC approaches targeting STn antigens by conjugating them to monomethyl auristatin E (MMAE) showed promising antitumor activity.(76,77) Immunotoxins such as diphtheria toxin fused to the single-chain variable fragment (scFv) 5F11 and pseudomonas toxin fused to scFv14.18—demonstrated specific killing of GD2+ tumor cells. However, immunogenicity of foreign toxin proteins has remained a major concern, and no GD2-directed immunotoxins are currently in human trials.(78) Regression of lung, breast, and bladder carcinomas in patient derived xenograph (PDX) models was demonstrated upon administration of BR96 sFv-PE40 Pseudomonas exotoxin A.(79) The first human study to obtain data on the safety and feasibility of 89Zr-DFO-HumAb-5B1 to image pancreatic tumors and other SLea-positive malignancies is ongoing (NCT02687230).
Vaccines
Targeting TACAs as an immunotherapeutic strategy with anticancer vaccines provides an appealing option for cancer treatment. Examples include vaccines targeting the mucin-related Tn, STn, and T antigens, the gangliosides GM2 and GD3, and the glycosphingolipid Globo-H.(80–82) Several carbohydrate-based vaccines have shown some promise and are presently undergoing clinical evaluation (Table 2). Strategies to overcome poor immunogenicity of glycan-based vaccines by displaying vaccine glycans in a multivalent context are currently being pursued. For example, a triantigenic vaccine containing Globo H, Ley, and Tn has been shown in animal models to elicit an immune response against each oligosaccharide antigen and it may result in recruiting both humoral and T cell-mediated immune responses against tumors in human patients.(83) Similarly, vaccination of small cell lung carcinoma patients with polysialic acid (PSA)-KLH or N-propionylated PSA-KLH (PrPSA-KLH) conjugates was tested in a clinical trial.(84)
Successful tumor immunotherapy might require the induction of cytotoxic T lymphocytes (CTLs) in addition to antibodies, which explains early attempts of developing a carbohydrate-based vaccine such as Theratope that elicits a B cell-mediated immune response, but does not seem to trigger a T cell-mediated immune response.(85) Another approach to overcome poor immunogenicity and lack of T cell engagement has been developed using anti-idiotype antibodies (Ab2) as surrogate antigens. Ab2 vaccines induce anti-GD2 immune responses and have several advantages over native gangliosides; as proteins, they induce T cell responses. Anti-idiotypic GD2 mAbs were tested in phase I trials.(86)
An alternative approach to develop T-dependent responses to carbohydrate Ags is the use of peptide or polypeptide surrogates of carbohydrates, which has also been carried out by our group in collaboration with the University of Pennsylvania. Peptides may functionally mimic carbohydrates and induce IgG and cellular immune responses, which the carbohydrate itself is usually unable to induce.(87–90) Carbohydrate-mimetic peptides (CMPs), unlike carbohydrate antigens, can prime for memory responses to TACAs, suggesting that CMPs facilitate cognate interactions between B cells and T cells. Antibodies induced by a CMP to the meningococcal group C capsular polysaccharide(91) were shown to be reactive with the LeY antigen and activated peptide-specific T helper type 1 (Th1) and type 2 (Th2) responses.(92,93) To induce sustained immunity against both LeY and GD2, CMP as a surrogate pan-immunogen that mimics both was developed. To test the feasibility of inducing proapoptotic antibodies reactive with LeY and GD2, advanced breast cancer subjects were immunized with the P10s-PADRE vaccine and limited clinical benefit was observed following vaccine treatment, which was not only caspase 3 dependent but also demonstrated synergy with chemotherapeutics(94) (NCT02229084 and NCT02264236).
Similarly, a DNA vaccine encoding a peptide isolated with the GD2-specific mAb 14G2a GD2, resulted in the induction of antibodies that exhibited CDC against GD2+ melanoma cells and inhibited growth of human melanoma cell xenografts. A study suggests that peptides mimicking the GD2 ganglioside inhibit tumor growth through antibody and/or CD4+ T cell-mediated mechanisms. DNA vaccination studies in mice showed that plasmids encoding peptides mimicking LeY induced LeY cross-reactive IgG2a Abs and mediated CMC.(95) Peptide mimetic of carbohydrate Ags encoded as DNA plasmids are novel immunogens providing a means to manipulate carbohydrate cross-reactive Th1 responses. This approach provides a way for development of messenger RNA (mRNA) vaccines targeting the immune response to glycans.
Adhesion Antagonists
Lectins such as C-type, Galectins, and Siglecs are important for adaptive immune responses. Examples of a lectin–ligand interaction (important for the homing and tissue recruitment of leukocytes and tumor cells) involve the C-type lectins, E-selectin, P-selectin, and L-selectin (CD62E, CD62P, and CD62L), and LeX or SLea that are the major ligands expressed on the surface of tumor cells.(96,97) During inflammation, selectins mediate the initial attachment of leukocytes to the endothelium during the process of leukocyte extravasation. In cancer, SLeX and SLea interactions with selectins regulate the metastatic cascade by forming emboli of cancer cells and platelets and favoring their arrest on endothelia.
We hypothesized that the tumor cell transmigration from the bloodstream to metastatic sites in analogy with lymphocyte rolling is mediated by interaction of selectins on endothelial cells and TACAs on the surface of tumor cells. Applying the concept of the functional equivalence of chemically dissimilar molecules such as carbohydrates and proteins sharing common surface topology, our study of administration of a monovalent peptide mimetic of SLea showed reduced neutrophil recruitment in vivo.(98–100) In a subsequent study, colonization of tumor cells expressing SLea was blocked by the peptide mimetic of this antigen and was completely abolished in E-selectin knockout mice.(101,102) Using combinatorial synthetic chemistry technology, this study allowed for delineating the positions of amino acids containing carboxyl groups that improved upon peptide mimicry and increased mAb binding. Developing reagents that are stereochemically equivalent to carbohydrate ligands for E-selectin that can effectively block lymphocyte rolling and tumor colonization in vivo might provide an effective treatment of the metastatic process and inflammatory conditions. Consequently, interference with selectin functions has become a potential therapeutic strategy, and these compounds are currently in clinical development. Glycomimetics' E-selectin inhibitor Uproleselan in combination with chemotherapy has been shown to improve survival in patients with acute myeloid leukemia (NCT03616470).(103)
Galectins are endogenous lectins recognizing galactose that allows for specific binding to carbohydrate epitopes, which can be shared by several T cell surface proteins. Galectins are expressed in cancer and stromal cells and mediate interactions between tumor cells and innate and adaptive immune cells. Galectin-3 binds to the cytotoxic T lymphocyte antigen 4 (CTLA-4) and lymphocyte activation gene 3 (LAG-3), while galectin-9 binds to T cell immunoglobulin and mucin-domain containing-3 (TIM-3).(104,105) Galectin-1 binding induces partial phosphorylation of TCRz and induces partial z-chain phosphorylation (pp21z) that cannot initiate downstream protein tyrosine phosphorylation, IL-2 production, or T cell proliferation.(106) Because of their immunosuppressive role, targeting galectins represents a potential therapeutic approach to restore antitumor immunity. Galectin antagonists in combination with chemotherapy, peptide vaccinations, or immune checkpoint inhibitors are currently in clinical trials for treatment of different cancers (Table 2).
The Siglec family of lectins comprises immunoglobulin-type lectins, which recognize predominantly sialic acid-containing glycans that are expressed in most white blood cells of the immune system and play critical roles in immune cell signaling.(107) For example, Siglec-9 plays a critical role in suppressing antigen-specific T cell responses in vitro and in vivo. Siglec-9 is coexpressed with several known inhibitory T cell receptors, for example, PD-1, CTLA-4, and TIM-3, in healthy individuals and melanoma patients.
A subset of Siglec-9 CD8 T effector cell engagement was associated with phosphorylation of the inhibitory protein tyrosine phosphatase SHP-1, but not SHP-2, resulting in suppressed TCR signaling and effector functions.(108) Siglec-15 is not expressed in normal tissue, but is upregulated in tumor cells and tumor-associated myeloid cells as well as M2 macrophages, leading to profound immunosuppression in the tumor microenvironment (TME). Siglec-15 expression is mutually exclusive with that of PD-L1, suggesting that a drug that alleviates Siglec-15–driven immunosuppression could be viable in patients with low PD-L1 expression to benefit from checkpoint blockade.(109) Many avenues to exploit sialic acid–Siglec interactions to advance cancer therapy are under investigation for advanced solid tumors (NCT03665285).
CAR-Modified T Cells and BsAbs
CARs and BsAbs are exciting developments for TACA-based immunotherapy. Adoptive transfer of CAR T cells is a promising immunotherapy strategy to treat cancer in an MHC-independent manner. CARs are genetically encoded artificial TCRs that combine the antigen specificity of an antibody with the machinery of T cell activation. CARs are generated by linking the scFv of an mAb with the TCRζ-chain transmembrane and cytoplasmic regions. The second- and third-generation CARs include additional signaling domains (CD27, 4-1BB, or OX40) aimed at improving proliferation, survival, and cytokine release from the cells. CAR immunotherapies have been shown to be highly effective in hematological malignancies (KYMRIAH and CARTA) and have been approved by the FDA. However, translation of CAR-T cell therapies from hematologic malignancies into solid tumors comes with challenges, including immunosuppressive TME.
CAR T cells based on mAbs targeting GD2 and LeY were demonstrated to be effective in eradicating leukemia and pancreatic cancer in mice(110) and delayed growth of myeloma xenografts.(111) Humanized Ab-based second generation of CARs targeting LeY coupled to the cytoplasmic domains of CD28 and the TCRz chain showed durable persistence.(112) An ongoing phase I clinical trial is now testing the safety and tolerability of using these CAR T cells in patients with advanced solid tumors presenting Ley surface expression (NCT03851146). A third generation of GD2-specific CAR-T cells has been developed and tested in a phase I clinical trial.(113) Built-in costimulatory domains such as CD28 and OX40 in T cells help to maintain the ability of cells to proliferate as well as to reduce T cell exhaustion and activation-induced cell death.(114) Other stimulatory molecules have been incorporated into GD2 CAR T cells, which enabled long-term persistence of the cells in human patients and led to improved clinical outcome.(115)
As an alternative to CAR T cells, BsAbs have shown great promise in anticancer therapy. BiAbs, similar to CAR T cells, do not require HLA for antigen presentation. The most common TACA-based BsAbs target GD2. BsAbs produced by conjugating anti-CD3 (OKT3) and anti-GD2 (3F8) antibodies recognize the tumor-associated ganglioside GD2, and the T cell receptor antigen CD3 can activate and redirect non-MHC-restricted cytotoxic activity.(116) More recent studies have substituted the 5F11-scFv with the higher-affinity hu3F8-scFv to form hu3F8-scBA. These BsAb-redirected T cells induced stronger T cell activation in vitro and more effectively suppressed NB xenograft growth in vivo compared with 5F11-scBA.(117) Bispecific anti-CD3 × anti-GD2 is tested in children with NBs and other GD2-expressing tumors (NCT02173093).
A direct comparison of GD2 BsAbs and CAR T cells found that BsAbs lead to longer survival of activated T cells. Furthermore, BsAb T cells provided more effective tumor protection in tumor models. The superiority of BsAb T cells could be partly attributed to the presence of CD4+ helper T cells, while the infused CAR T cells were almost exclusively CD8+ T cells. This incomplete benefit may reflect the difficulties of sustaining human T cell function and trafficking in a xenogeneic environment, which may represent the limitations of even the third-generation constructs that cannot completely recapitulate the temporo-spatial features of costimulatory events required to physiologically sustain T cell activation.(118)
Concluding Remarks
Advances in molecular targeted therapy and immune checkpoint inhibition have led to unprecedented improvement in overall survival of patients with cancer. Despite these improvements, there is an unmet need for novel approaches for next-generation, antitumor immune modulatory drugs. There is increasing evidence that altered glycans that are active players throughout cancer development and progression can be targeted for effective therapies.
While the results from the studies conducted at the Wistar Institute decades ago using carbohydrate-binding mAbs have not been recognized for their clinical potential, these early studies provided the conceptual framework for the current advances that will likely further progress in development of agents based on recognition of TACAs.
Currently, diverse and innovative approaches targeting cancer-associated glycans, such as mAbs, BsAbs, and glycan-specific CAR-T cells; carbohydrates and carbohydrate analog-based vaccines; adhesion antagonists; and small molecules with potential clinical application are tested. A structure-based design of mAbs with improved potency and selectivity for tumors over normal tissues may now be a tangible option, especially in systems where detailed, three-dimensional structural information is available, such as the Lewis histo-blood group and related TACAs. Such innovative strategies are likely to overcome many current limitations in the diagnosis and treatment of cancer patients. As discussed in this review, the strategies that exploit aberrant glycosylation of cancer cells can provide therapeutic options and act synergistically with current targeted approaches and immunotherapy approaches, improving their specificity and efficacy.
Author Disclosure Statement
No competing financial interests exist.
Funding Information
No funding was received for this article.
References
- 1. Kieber-Emmons A, Monzavi-Karbassi B, and Kieber-Emmons T: Antigens: carbohydrates II. In: eLS. 2020, pp. 1–29 . DOI: 10.1002/9780470015902.a0000500.pub2 [DOI] [Google Scholar]
- 2. Hakomori S, and Zhang Y: Glycosphingolipid antigens and cancer therapy. Chem Biol 1997;4:97–104 [DOI] [PubMed] [Google Scholar]
- 3. Koprowski H, Steplewski Z, Herlyn D, and Herlyn M: Study of antibodies against human melanoma produced by somatic cell hybrids. Proc Natl Acad Sci U S A 1978;75:3405–3409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Koprowski H, Steplewski Z, Mitchell K, Herlyn M, Herlyn D, and Fuhrer P: Colorectal carcinoma antigens detected by hybridoma antibodies. Somatic Cell Genet 1979;5:957–971 [DOI] [PubMed] [Google Scholar]
- 5. Herlyn M, Steplewski Z, Herlyn D, and Koprowski H: Colorectal carcinoma-specific antigen: detection by means of monoclonal antibodies. Proc Natl Acad Sci U S A 1979;76:1438–1442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Magnani JL, Brockhaus M, Smith DF, Ginsburg V, Blaszczyk M, Mitchell KF, Steplewski Z, and Koprowski H: A monosialoganglioside is a monoclonal antibody-defined antigen of colon carcinoma. Science 1981;212:55–56 [DOI] [PubMed] [Google Scholar]
- 7. Magnani JL, Nilsson B, Brockhaus M, Zopf D, Steplewski Z, Koprowski H, and Ginsburg V: A monoclonal antibody-defined antigen associated with gastrointestinal cancer is a ganglioside containing sialylated lacto-N-fucopentaose II. J Biol Chem 1982;257:14365–14369 [PubMed] [Google Scholar]
- 8. Engle DD, Tiriac H, Rivera KD, Pommier A, Whalen S, Oni TE, Alagesan B, Lee EJ, Yao MA, Lucito MS, Spielman B, Da Silva B, Schoepfer C, Wright K, Creighton B, Afinowicz L, Yu KH, Grützmann R, Aust D, Gimotty PA, Pollard KS, Hruban RH, Goggins MG, Pilarsky C, Park Y, Pappin DJ, Hollingsworth MA, and Tuveson DA: The glycan CA19-9 promotes pancreatitis and pancreatic cancer in mice. Science 2019;364:1156–1162 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Brockhaus M, Magnani JL, Herlyn M, Blaszczyk M, Steplewski Z, Koprowski H, and Ginsburg V: Monoclonal antibodies directed against the sugar sequence of lacto-N-fucopentaose III are obtained from mice immunized with human tumors. Arch Biochem Biophys 1982;217:647–651 [DOI] [PubMed] [Google Scholar]
- 10. Brockhaus M, Magnani JL, Blaszczyk M, Steplewski Z, Koprowski H, Karlsson KA, Larson G, and Ginsburg V: Monoclonal antibodies directed against the human Leb blood group antigen. J Biol Chem 1981;256:13223–13225 [PubMed] [Google Scholar]
- 11. Hansson GC, Karlsson KA, Larson G, McKibbin JM, Blaszczyk M, Herlyn M, Steplewski Z, and Koprowski H: Mouse monoclonal antibodies against human cancer cell lines with specificities for blood group and related antigens. Characterization by antibody binding to glycosphingolipids in a chromatogram binding assay. J Biol Chem 1983;258:4091–4097 [PubMed] [Google Scholar]
- 12. Blaszczyk M, Hansson GC, Karlsson KA, Larson G, Stromberg N, Thurin J, Herlyn M, Steplewski Z, and Koprowski H: Lewis blood group antigens defined by monoclonal anti-colon carcinoma antibodies. Arch Biochem Biophys 1984;233:161–168 [DOI] [PubMed] [Google Scholar]
- 13. Blaszczyk M, Ross AH, Ernst CS, Marchisio M, Atkinson BF, Pak KY, Steplewski Z, and Koprowski H: A fetal glycolipid expressed on adenocarcinomas of the colon. Int J Cancer 1984;33:313–318 [DOI] [PubMed] [Google Scholar]
- 14. Olding LB, Ahren C, Thurin J, Karlsson KA, Svalander C, and Koprowski H: Gastrointestinal carcinoma-associated antigen detected by a monoclonal antibody in dysplasia and adenocarcinoma associated with chronic ulcerative colitis. Int J Cancer 1985;36:131–136 [DOI] [PubMed] [Google Scholar]
- 15. Olding LB, Thurin J, Svalander C, and Koprowski H: Expression of gastrointestinal carcinoma-associated antigen (GICA) detected in human fetal tissues by monoclonal antibody NS-19-9. Int J Cancer 1984;34:187–192 [DOI] [PubMed] [Google Scholar]
- 16. Blaszczyk M, Pak KY, Herlyn M, Sears HF, and Steplewski Z: Characterization of Lewis antigens in normal colon and gastrointestinal adenocarcinomas. Proc Natl Acad Sci U S A 1985;82:3552–3556 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Okada Y, Arima T, Togawa K, Nagashima H, Jinno K, Moriwaki S, Kunitomo T, Thurin J, and Koprowski H: Neoexpression of ABH and Lewis blood group antigens in human hepatocellular carcinomas. J Natl Cancer Inst 1987;78:19–28 [DOI] [PubMed] [Google Scholar]
- 18. Thurin J, Brodin T, Bechtel B, Jovall PA, Karlsson H, Strömberg N, Teneberg S, Sjögren HO, and Karlsson KA: Novel isoglobo-neolacto-series hybrid glycolipid detected by a monoclonal antibody is a rat colon tumor-associated antigen. Biochim Biophys Acta 1989;1002:267–272 [DOI] [PubMed] [Google Scholar]
- 19. Rodeck U, Herlyn M, Leander K, Borlinghaus P, and Koprowski H: A mucin containing the X, Y, and H type 2 carbohydrate determinants is shed by carcinoma cells. Hybridoma 1987;6:389–401 [DOI] [PubMed] [Google Scholar]
- 20. Blaszczyk M, Pak KY, Herlyn M, Lindgren J, Pessano S, Steplewski Z, and Koprowski H: Characterization of gastrointestinal tumor-associated carcinoembryonic antigen-related antigens defined by monoclonal antibodies. Cancer Res 1984;44:245–253 [PubMed] [Google Scholar]
- 21. Solter D, and Knowles BB: Monoclonal antibody defining a stage-specific mouse embryonic antigen (SSEA-1). Proc Natl Acad Sci U S A 1978;75:5565–5569 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kannagi R, Cochran NA, Ishigami F, Hakomori S, Andrews PW, Knowles BB, and Solter D: Stage-specific embryonic antigens (SSEA-3 and -4) are epitopes of a unique globo-series ganglioside isolated from human teratocarcinoma cells. EMBO J 1983;2:2355–2361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Atkinson BF, Ernst CS, Herlyn M, Steplewski Z, Sears HF, and Koprowski H: Gastrointestinal cancer-associated antigen in immunoperoxidase assay. Cancer Res 1982;42:4820–4823 [PubMed] [Google Scholar]
- 24. Atkinson B, Ernst CS, Ghrist BF, Herlyn M, Blaszczyk M, Ross AH, Herlyn D, Steplewski Z, and Koprowski H: Identification of melanoma-associated antigens using fixed tissue screening of antibodies. Cancer Res 1984;44:2577–2581 [PubMed] [Google Scholar]
- 25. Ernst C, Atkinson B, Wysocka M, Blaszczyk M, Herlyn M, Sears H, Steplewski Z, and Koprowski H: Monoclonal antibody localization of Lewis antigens in fixed tissue. Lab Invest 1984;50:394–400 [PubMed] [Google Scholar]
- 26. Ernst C, Thurin J, Atkinson B, Wurzel H, Herlyn M, Stromberg N, Civin C, and Koprowski H: Monoclonal antibody localization of A and B isoantigens in normal and malignant fixed human tissues. Am J Pathol 1984;117:451–461 [PMC free article] [PubMed] [Google Scholar]
- 27. Herlyn M, Steplewski Z, Herlyn D, Clark WH Jr., Ross AH, Blaszczyk M, Pak KY, and Koprowski H: Production and characterization of monoclonal antibodies against human malignant melanoma. Cancer Invest 1983;1:215–224 [DOI] [PubMed] [Google Scholar]
- 28. Thurin J, Thurin M, Kimoto Y, Herlyn M, Lubeck MD, Elder DE, Smereczynska M, Karlsson KA, Clark WM Jr., Steplewski Z, and Koprowski H: Monoclonal antibody-defined correlations in melanoma between levels of GD2 and GD3 antigens and antibody-mediated cytotoxicity. Cancer Res 1987;47:1229–1233 [PubMed] [Google Scholar]
- 29. Elder DE, Rodeck U, Thurin J, Cardillo F, Clark WH, Stewart R, and Herlyn M: Antigenic profile of tumor progression stages in human melanocytic nevi and melanomas. Cancer Res 1989;49:5091–5096 [PubMed] [Google Scholar]
- 30. Herlyn M, Thurin J, Balaban G, Bennicelli JL, Herlyn D, Elder DE, Bondi E, Guerry D, Nowell P, Clark WH, and Koprowski H: Characteristics of cultured human melanocytes isolated from different stages of tumor progression. Cancer Res 1985;45:5670–5676 [PubMed] [Google Scholar]
- 31. Lichtin AE, Guerry D, Elder DE, Hamilton R, LaRossa D, Herlyn D, Iliopoulos D, Thurin J, and Steplewski Z: A phase I study of monoclonal antibody therapy in disseminated melanoma. In: Proceedings of the 8th International Pigment Cell Conference, Tucson, AZ, 1986 [Google Scholar]
- 32. Blaszczyk-Thurin M, Sarnesto A, Thurin J, Hindsgaul O, and Koprowski H: Biosynthetic pathways for the Leb and Y glycolipids in the gastric carcinoma cell line KATO III as analyzed by a novel assay. Biochem Biophys Res Commun 1988;151:100–108 [DOI] [PubMed] [Google Scholar]
- 33. Thurin J, Thurin M, Herlyn M, Elder DE, Steplewski Z, Clark WH Jr., and Koprowski H: GD2 ganglioside biosynthesis is a distinct biochemical event in human melanoma tumor progression. FEBS Lett 1986;208:17–22 [DOI] [PubMed] [Google Scholar]
- 34. Sarnesto A, Köhlin T, Hindsgaul O, Thurin J, and Blaszczyk-Thurin M: Purification of the secretor-type beta-galactoside alpha 1——2-fucosyltransferase from human serum. J Biol Chem 1992;267:2737–2744 [PubMed] [Google Scholar]
- 35. Sarnesto A, Köhlin T, Thurin J, and Blaszczyk-Thurin M: Purification of H gene-encoded beta-galactoside alpha 1——2 fucosyltransferase from human serum. J Biol Chem 1990;265:15067–15075 [PubMed] [Google Scholar]
- 36. Thurin J, and Blaszczyk-Thurin M: Porcine submaxillary gland GDP-L-fucose: beta-D-galactoside alpha-2-L-fucosyltransferase is likely a counterpart of the human Secretor gene-encoded blood group transferase. J Biol Chem 1995;270:26577–26580 [DOI] [PubMed] [Google Scholar]
- 37. Sun J, Thurin J, Cooper HS, Wang P, Mackiewicz M, Steplewski Z, and Blaszczyk-Thurin M: Elevated expression of H type GDP-L-fucose:beta-D-galactoside alpha-2-L-fucosyltransferase is associated with human colon adenocarcinoma progression. Proc Natl Acad Sci U S A 1995;92:5724–5728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Hindsgaul O, Kaur KJ, Srivastava G, Blaszczyk-Thurin M, Crawley SC, Heerze LD, and Palcic MM: Evaluation of deoxygenated oligosaccharide acceptor analogs as specific inhibitors of glycosyltransferases. J Biol Chem 1991;266:17858–17862 [PubMed] [Google Scholar]
- 39. Bechtel B, Wand AJ, Wroblewski K, Koprowski H, and Thurin J: Conformational analysis of the tumor-associated carbohydrate antigen 19-9 and its Lea blood group antigen component as related to the specificity of monoclonal antibody CO19-9. J Biol Chem 1990;265:2028–2037 [PubMed] [Google Scholar]
- 40. Falk KE, Karlsson KA, Larson G, Thurin J, Blaszczyk M, Steplewski Z, and Koprowski H: Mass spectrometry of a human tumor glycolipid antigen being defined by mouse monoclonal antibody NS-19-9. Biochem Biophys Res Commun 1983;110:383–391 [DOI] [PubMed] [Google Scholar]
- 41. Thurin J, Herlyn M, Hindsgaul O, Strömberg N, Karlsson KA, Elder D, Steplewski Z, and Koprowski H: Proton NMR and fast-atom bombardment mass spectrometry analysis of the melanoma-associated ganglioside 9-O-acetyl-GD3. J Biol Chem 1985;260:14556–14563 [PubMed] [Google Scholar]
- 42. Blaszczyk-Thurin M, Thurin J, Hindsgaul O, Karlsson KA, Steplewski Z, and Koprowski H: Y and blood group B type 2 glycolipid antigens accumulate in a human gastric carcinoma cell line as detected by monoclonal antibody. Isolation and characterization by mass spectrometry and NMR spectroscopy. J Biol Chem 1987;262:372–379 [PubMed] [Google Scholar]
- 43. Blaszczyk-Thurin M, Murali R, Westerink MA, Steplewski Z, Co MS, and Kieber-Emmons T: Molecular recognition of the Lewis Y antigen by monoclonal antibodies. Protein Eng 1996;9:447–459 [DOI] [PubMed] [Google Scholar]
- 44. Murali R, and Kieber-Emmons T: Molecular recognition of a peptide mimic of the Lewis Y antigen by an anti-Lewis Y antibody. J Mol Recognit 1997;10:269–276 [DOI] [PubMed] [Google Scholar]
- 45. Saha S, Pashov A, Siegel ER, Murali R, and Kieber-Emmons T: Defining the recognition elements of Lewis Y-reactive antibodies. PLoS One 2014;9:e104208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Saha S, Murali R, Pashov A, and Kieber-Emmons T: The potential role of solvation in antibody recognition of the Lewis Y antigen. Monoclon Antib Immunodiagn Immunother 2015;34:295–302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Ramsland PA, Farrugia W, Bradford TM, Mark Hogarth P, and Scott AM: Structural convergence of antibody binding of carbohydrate determinants in Lewis Y tumor antigens. J Mol Biol 2004;340:809–818 [DOI] [PubMed] [Google Scholar]
- 48. Dingjan T, Spendlove I, Durrant LG, Scott AM, Yuriev E, and Ramsland PA: Structural biology of antibody recognition of carbohydrate epitopes and potential uses for targeted cancer immunotherapies. Mol Immunol 2015;67:75–88 [DOI] [PubMed] [Google Scholar]
- 49. Kirwan A, Utratna M, O'Dwyer ME, Joshi L, and Kilcoyne M: Glycosylation-based serum biomarkers for cancer diagnostics and prognostics. Biomed Res Int 2015;2015:490531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Herlyn M, Sears HF, Steplewski Z, and Koprowski H: Monoclonal antibody detection of a circulating tumor-associated antigen. I. Presence of antigen in sera of patients with colorectal, gastric, and pancreatic carcinoma. J Clin Immunol 1982;2:135–140 [DOI] [PubMed] [Google Scholar]
- 51. Herlyn M, Blaszczyk M, Sears HF, Verrill H, Lindgren J, Colcher D, Steplewski Z, Schlom J, and Koprowski H: Detection of carcinoembryonic antigen and related antigens in sera of patients with gastrointestinal tumors using monoclonal antibodies in double-determinant radioimmunoassays. Hybridoma 1983;2:329–339 [DOI] [PubMed] [Google Scholar]
- 52. Steplewski Z, Herlyn M, Blaszczyk M, and Koprowski H: A simple procedure for determining Lewis phenotypes in human saliva. J Immunol Methods 1983;62:73–78 [DOI] [PubMed] [Google Scholar]
- 53. Pak KY, Blaszczyk M, Herlyn M, Steplewski Z, and Koprowski H: Identification and isolation of Lewis blood group antigens from human saliva using monoclonal antibodies. Hybridoma 1984;3:1–10 [DOI] [PubMed] [Google Scholar]
- 54. Herlyn M, Blaszczyk M, Bennicelli J, Sears HF, Ernst C, Ross AH, and Koprowski H: Selection of monoclonal antibodies detecting serodiagnostic human tumor markers. J Immunol Methods 1985;80:107–116 [DOI] [PubMed] [Google Scholar]
- 55. Brockhaus M, Wysocka M, Magnani JL, Steplewski Z, Koprowski H, and Ginsburg V: Normal salivary mucin contains the gastrointestinal cancer-associated antigen detected by monoclonal antibody 19-9 in the serum mucin of patients. Vox Sang 1985;48:34–38 [DOI] [PubMed] [Google Scholar]
- 56. Vestergaard EM, Hein HO, Meyer H, Grunnet N, Jørgensen J, Wolf H, and Orntoft TF: Reference values and biological variation for tumor marker CA 19-9 in serum for different Lewis and secretor genotypes and evaluation of secretor and Lewis genotyping in a Caucasian population. Clin Chem 1999;45:54–61 [PubMed] [Google Scholar]
- 57. Safi F, Schlosser W, Kolb G, and Beger HG: Diagnostic value of CA 19-9 in patients with pancreatic cancer and nonspecific gastrointestinal symptoms. J Gastrointest Surg 1997;1:106–112 [DOI] [PubMed] [Google Scholar]
- 58. Koprowski H, Brockhaus M, Blaszczyk M, Magnani J, Steplewski Z, and Ginsburg V: Lewis blood-type may affect the incidence of gastrointestinal cancer. Lancet 1982;1:1332–1333 [DOI] [PubMed] [Google Scholar]
- 59. Rodrigues Mantuano N, Natoli M, Zippelius A, and Läubli H: Tumor-associated carbohydrates and immunomodulatory lectins as targets for cancer immunotherapy. J Immunother Cancer 2020;8:e001222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Mereiter S, Balmaña M, Campos D, Gomes J, and Reis CA: Glycosylation in the era of cancer-targeted therapy: where are we heading? Cancer Cell 2019;36:6–16 [DOI] [PubMed] [Google Scholar]
- 61. Gupta S, McDonald JD, Ayabe RI, Khan TM, Gamble LA, Sinha S, Hannah C, Blakely AM, Davis JL, and Hernandez JM: Targeting CA 19-9 with a humanized monoclonal antibody at the time of surgery may decrease recurrence rates for patients undergoing resections for pancreatic cancer, cholangiocarcinoma and metastatic colorectal cancer. J Gastrointest Oncol 2020;11:231–235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Ragupathi G, Damani P, Srivastava G, Srivastava O, Sucheck SJ, Ichikawa Y, and Livingston PO: Synthesis of sialyl Lewis(a) (sLe (a), CA19-9) and construction of an immunogenic sLe(a) vaccine. Cancer Immunol Immunother 2009;58:1397–1405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Sawada R, Sun SM, Wu X, Hong F, Ragupathi G, Livingston PO, and Scholz WW: Human monoclonal antibodies to sialyl-Lewis (CA19.9) with potent CDC, ADCC, and antitumor activity. Clin Cancer Res 2011;17:1024–1032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Weitzenfeld P, Bournazos S, and Ravetch JV: Antibodies targeting sialyl Lewis A mediate tumor clearance through distinct effector pathways. J Clin Invest 2019;129:3952–3962 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Steplewski Z, Spira G, Blaszczyk M, Lubeck MD, Radbruch A, Illges H, Herlyn D, Rajewsky K, and Scharff M: Isolation and characterization of anti-monosialoganglioside monoclonal antibody 19-9 class-switch variants. Proc Natl Acad Sci U S A 1985;82:8653–8657 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Steplewski Z, Blaszczyk-Thurin M, Lubeck M, Loibner H, Scholz D, and Koprowski H: Oligosaccharide Y specific monoclonal antibody and its isotype switch variants. Hybridoma 1990;9:201–210 [DOI] [PubMed] [Google Scholar]
- 67. Mujoo K, Kipps TJ, Yang HM, Cheresh DA, Wargalla U, Sander DJ, and Reisfeld RA: Functional properties and effect on growth suppression of human neuroblastoma tumors by isotype switch variants of monoclonal antiganglioside GD2 antibody 14.18. Cancer Res 1989;49:2857–2861 [PubMed] [Google Scholar]
- 68. Gillies SD, Lo KM, and Wesolowski J: High-level expression of chimeric antibodies using adapted cDNA variable region cassettes. J Immunol Methods 1989;125:191–202 [DOI] [PubMed] [Google Scholar]
- 69. Ozkaynak MF, Sondel PM, Krailo MD, Gan J, Javorsky B, Reisfeld RA, Matthay KK, Reaman GH, and Seeger RC: Phase I study of chimeric human/murine anti-ganglioside G(D2) monoclonal antibody (ch14.18) with granulocyte-macrophage colony-stimulating factor in children with neuroblastoma immediately after hematopoietic stem-cell transplantation: a Children's Cancer Group Study. J Clin Oncol 2000;18:4077–4085 [DOI] [PubMed] [Google Scholar]
- 70. Scott AM, Geleick D, Rubira M, Clarke K, Nice EC, Smyth FE, Stockert E, Richards EC, Carr FJ, Harris WJ, Armour KL, Rood J, Kypridis A, Kronina V, Murphy R, Lee FT, Liu Z, Kitamura K, Ritter G, Laughton K, Hoffman E, Burgess AW, and Old LJ: Construction, production, and characterization of humanized anti-Lewis Y monoclonal antibody 3S193 for targeted immunotherapy of solid tumors. Cancer Res 2000;60:3254–3261 [PubMed] [Google Scholar]
- 71. Smaletz O, Diz MD, do Carmo CC, Sabbaga J, Cunha-Junior GF, Azevedo SJ, Maluf FC, Barrios CH, Costa RL, Fontana AG, Madrigal V, Wainstein AJ, Yeda FP, Alves VA, Moro AM, Blasbalg R, Scott AM, and Hoffman EW: A phase II trial with anti-Lewis-Y monoclonal antibody (hu3S193) for the treatment of platinum resistant/refractory ovarian, fallopian tube and primary peritoneal carcinoma. Gynecol Oncol 2015;138:272–277 [DOI] [PubMed] [Google Scholar]
- 72. Scott AM, Tebbutt N, Lee FT, Cavicchiolo T, Liu Z, Gill S, Poon AM, Hopkins W, Smyth FE, Murone C, MacGregor D, Papenfuss AT, Chappell B, Saunder TH, Brechbiel MW, Davis ID, Murphy R, Chong G, Hoffman EW, and Old LJ: A phase I biodistribution and pharmacokinetic trial of humanized monoclonal antibody Hu3s193 in patients with advanced epithelial cancers that express the Lewis-Y antigen. Clin Cancer Res 2007;13:3286–3292 [DOI] [PubMed] [Google Scholar]
- 73. Krug LM, Milton DT, Jungbluth AA, Chen LC, Quaia E, Pandit-Taskar N, Nagel A, Jones J, Kris MG, Finn R, Smith-Jones P, Scott AM, Old L, and Divgi C: Targeting Lewis Y (Le(y)) in small cell lung cancer with a humanized monoclonal antibody, hu3S193: a pilot trial testing two dose levels. J Thorac Oncol 2007;2:947–952 [DOI] [PubMed] [Google Scholar]
- 74. Ross HJ, Hart LL, Swanson PM, Rarick MU, Figlin RA, Jacobs AD, McCune DE, Rosenberg AH, Baron AD, Grove LE, Thorn MD, Miller DM, Drachman JG, and Rudin CM: A randomized, multicenter study to determine the safety and efficacy of the immunoconjugate SGN-15 plus docetaxel for the treatment of non-small cell lung carcinoma. Lung Cancer 2006;54:69–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Tolcher AW, Sugarman S, Gelmon KA, Cohen R, Saleh M, Isaacs C, Young L, Healey D, Onetto N, and Slichenmyer W: Randomized phase II study of BR96-doxorubicin conjugate in patients with metastatic breast cancer. J Clin Oncol 1999;17:478–484 [DOI] [PubMed] [Google Scholar]
- 76. Prendergast JM, Galvao da Silva AP, Eavarone DA, Ghaderi D, Zhang M, Brady D, Wicks J, DeSander J, Behrens J, and Rueda BR: Novel anti-Sialyl-Tn monoclonal antibodies and antibody-drug conjugates demonstrate tumor specificity and anti-tumor activity. MAbs 2017;9:615–627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Tivadar ST, McIntosh RS, Chua JX, Moss R, Parsons T, Zaitoun AM, Madhusudan S, Durrant LG, and Vankemmelbeke M: Monoclonal antibody targeting Sialyl-di-Lewis(a)-containing internalizing and noninternalizing glycoproteins with cancer immunotherapy development potential. Mol Cancer Ther 2020;19:790–801 [DOI] [PubMed] [Google Scholar]
- 78. Thomas PB, Delatte SJ, Sutphin A, Frankel AE, and Tagge EP: Effective targeted cytotoxicity of neuroblastoma cells. J Pediatr Surg 2002;37:539–544 [DOI] [PubMed] [Google Scholar]
- 79. Siegall CB: Targeted therapy of carcinomas using BR96 sFv-PE40, a single-chain immunotoxin that binds to the Le(y) antigen. Semin Cancer Biol 1995;6:289–295 [DOI] [PubMed] [Google Scholar]
- 80. Danielczyk A, Stahn R, Faulstich D, Löffler A, Märten A, Karsten U, and Goletz S: PankoMab: a potent new generation anti-tumour MUC1 antibody. Cancer Immunol Immunother 2006;55:1337–1347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Sabbatini PJ, Kudryashov V, Ragupathi G, Danishefsky SJ, Livingston PO, Bornmann W, Spassova M, Zatorski A, Spriggs D, Aghajanian C, Soignet S, Peyton M, O'Flaherty C, Curtin J, and Lloyd KO: Immunization of ovarian cancer patients with a synthetic Lewis(y)-protein conjugate vaccine: a phase 1 trial. Int J Cancer 2000;87:79–85 [PubMed] [Google Scholar]
- 82. Krug LM, Ragupathi G, Hood C, Kris MG, Miller VA, Allen JR, Keding SJ, Danishefsky SJ, Gomez J, Tyson L, Pizzo B, Baez V, and Livingston PO: Vaccination of patients with small-cell lung cancer with synthetic fucosyl GM-1 conjugated to keyhole limpet hemocyanin. Clin Cancer Res 2004;10:6094–6100 [DOI] [PubMed] [Google Scholar]
- 83. Ragupathi G, Coltart DM, Williams LJ, Koide F, Kagan E, Allen J, Harris C, Glunz PW, Livingston PO, and Danishefsky SJ: On the power of chemical synthesis: immunological evaluation of models for multiantigenic carbohydrate-based cancer vaccines. Proc Natl Acad Sci U S A 2002;99:13699–13704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Krug LM, Ragupathi G, Ng KK, Hood C, Jennings HJ, Guo Z, Kris MG, Miller V, Pizzo B, Tyson L, Baez V, and Livingston PO: Vaccination of small cell lung cancer patients with polysialic acid or N-propionylated polysialic acid conjugated to keyhole limpet hemocyanin. Clin Cancer Res 2004;10:916–923 [DOI] [PubMed] [Google Scholar]
- 85. Holmberg LA, and Sandmaier BM: Vaccination with Theratope (STn-KLH) as treatment for breast cancer. Expert Rev Vaccines 2004;3:655–663 [DOI] [PubMed] [Google Scholar]
- 86. Foon KA, Sen G, Hutchins L, Kashala OL, Baral R, Banerjee M, Chakraborty M, Garrison J, Reisfeld RA, and Bhattacharya-Chatterjee M: Antibody responses in melanoma patients immunized with an anti-idiotype antibody mimicking disialoganglioside GD2. Clin Cancer Res 1998;4:1117–1124 [PubMed] [Google Scholar]
- 87. Steplewski Z, Thurin M, and Kieber-Emmons T: Antibodies: at the nexus of antigens and cancer vaccines. J Infect Dis 2015;212(Suppl 1):S59–S66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Cunto-Amesty G, Luo P, Monzavi-Karbassi B, Lees A, and Kieber-Emmons T: Exploiting molecular mimicry to broaden the immune response to carbohydrate antigens for vaccine development. Vaccine 2001;19:2361–2368 [DOI] [PubMed] [Google Scholar]
- 89. Kieber-Emmons T, Monzavi-Karbassi B, Hutchins LF, Pennisi A, and Makhoul I: Harnessing benefit from targeting tumor associated carbohydrate antigens. Hum Vaccin Immunother 2017;13:323–331 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Kieber-Emmons T, Luo P, Qiu J, Chang TY, O I, Blaszczyk-Thurin M, and Steplewski Z: Vaccination with carbohydrate peptide mimotopes promotes anti-tumor responses. Nat Biotechnol 1999;17:660–665 [DOI] [PubMed] [Google Scholar]
- 91. Westerink MA, Giardina PC, Apicella MA, and Kieber-Emmons T: Peptide mimicry of the meningococcal group C capsular polysaccharide. Proc Natl Acad Sci U S A 1995;92:4021–4025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Luo P, Canziani G, Cunto-Amesty G, and Kieber-Emmons T: A molecular basis for functional peptide mimicry of a carbohydrate antigen. J Biol Chem 2000;275:16146–16154 [DOI] [PubMed] [Google Scholar]
- 93. Wondimu A, Zhang T, Kieber-Emmons T, Gimotty P, Sproesser K, Somasundaram R, Ferrone S, Tsao CY, and Herlyn D: Peptides mimicking GD2 ganglioside elicit cellular, humoral and tumor-protective immune responses in mice. Cancer Immunol Immunother 2008;57:1079–1089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Hutchins LF, Makhoul I, Emanuel PD, Pennisi A, Siegel ER, Jousheghany F, Guo X, Pashov AD, Monzavi-Karbassi B, and Kieber-Emmons T: Targeting tumor-associated carbohydrate antigens: a phase I study of a carbohydrate mimetic-peptide vaccine in stage IV breast cancer subjects. Oncotarget 2017;8:99161–99178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Kieber-Emmons T, Monzavi-Karbassi B, Wang B, Luo P, and Weiner DB: Cutting edge: DNA immunization with minigenes of carbohydrate mimotopes induce functional anti-carbohydrate antibody response. J Immunol 2000;165:623–627 [DOI] [PubMed] [Google Scholar]
- 96. McEver RP: Selectin-carbohydrate interactions during inflammation and metastasis. Glycoconj J 1997;14:585–591 [DOI] [PubMed] [Google Scholar]
- 97. Rosen SD, and Bertozzi CR: The selectins and their ligands. Curr Opin Cell Biol 1994;6:663–673 [DOI] [PubMed] [Google Scholar]
- 98. Thurin M, and Kieber-Emmons T: SA-Lea and tumor metastasis: the old prediction and recent findings. Hybrid Hybridomics 2002;21:111–116 [DOI] [PubMed] [Google Scholar]
- 99. O I, Kieber-Emmons T, Otvos L, and Blaszczyk-Thurin M: Peptide mimicking sialyl-Lewis(a) with anti-inflammatory activity. Biochem Biophys Res Commun 2000;268:106–111 [DOI] [PubMed] [Google Scholar]
- 100. O I, Kieber-Emmons T, Otvos L, Jr., and Blaszczyk-Thurin M: Peptides mimicking sialyl-Lewis A isolated from a random peptide library and peptide array. Ann N Y Acad Sci 1999;886:276–279 [DOI] [PubMed] [Google Scholar]
- 101. O I, Otvos L, Kieber-Emmons T, and Blaszczyk-Thurin M: Role of SA-Le(a) and E-selectin in metastasis assessed with peptide antagonist. Peptides 2002;23:999–1010 [DOI] [PubMed] [Google Scholar]
- 102. Monzavi-Karbassi B, Whitehead TL, Jousheghany F, Artaud C, Hennings L, Shaaf S, Slaughter A, Korourian S, Kelly T, Blaszczyk-Thurin M, and Kieber-Emmons T: Deficiency in surface expression of E-selectin ligand promotes lung colonization in a mouse model of breast cancer. Int J Cancer 2005;117:398–408 [DOI] [PubMed] [Google Scholar]
- 103. Muz B, Abdelghafer A, Markovic M, Yavner J, Melam A, Salama NN, and Azab AK: Targeting E-selectin to Tackle Cancer Using Uproleselan. Cancers (Basel) 2021;13:335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Kouo T, Huang L, Pucsek AB, Cao M, Solt S, Armstrong T, and Jaffee E: Galectin-3 shapes antitumor immune responses by suppressing CD8+ T cells via LAG-3 and inhibiting expansion of plasmacytoid dendritic cells. Cancer Immunol Res 2015;3:412–423 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, Zheng XX, Strom TB, and Kuchroo VK: The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 2005;6:1245–1252 [DOI] [PubMed] [Google Scholar]
- 106. Chung CD, Patel VP, Moran M, Lewis LA, and Miceli MC: Galectin-1 induces partial TCR zeta-chain phosphorylation and antagonizes processive TCR signal transduction. J Immunol 2000;165:3722–3729 [DOI] [PubMed] [Google Scholar]
- 107. Duan S, and Paulson JC: Siglecs as immune cell checkpoints in disease. Annu Rev Immunol 2020;38:365–395 [DOI] [PubMed] [Google Scholar]
- 108. Haas Q, Boligan KF, Jandus C, Schneider C, Simillion C, Stanczak MA, Haubitz M, Seyed Jafari SM, Zippelius A, Baerlocher GM, Läubli H, Hunger RE, Romero P, Simon HU, and von Gunten S: Siglec-9 regulates an effector memory CD8(+) T-cell subset that congregates in the melanoma tumor microenvironment. Cancer Immunol Res 2019;7:707–718 [DOI] [PubMed] [Google Scholar]
- 109. Wang J, Sun J, Liu LN, Flies DB, Nie X, Toki M, Zhang J, Song C, Zarr M, Zhou X, Han X, Archer KA, O'Neill T, Herbst RS, Boto AN, Sanmamed MF, Langermann S, Rimm DL, and Chen L: Siglec-15 as an immune suppressor and potential target for normalization cancer immunotherapy. Nat Med 2019;25:656–666 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Steentoft C, Migliorini D, King TR, Mandel U, June CH, and Posey AD Jr.: Glycan-directed CAR-T cells. Glycobiology 2018;28:656–669 [DOI] [PubMed] [Google Scholar]
- 111. Peinert S, Prince HM, Guru PM, Kershaw MH, Smyth MJ, Trapani JA, Gambell P, Harrison S, Scott AM, Smyth FE, Darcy PK, Tainton K, Neeson P, Ritchie DS, and Hönemann D: Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther 2010;17:678–686 [DOI] [PubMed] [Google Scholar]
- 112. Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K, Chen K, Shin M, Wall DM, Hönemann D, Gambell P, Westerman DA, Haurat J, Westwood JA, Scott AM, Kravets L, Dickinson M, Trapani JA, Smyth MJ, Darcy PK, Kershaw MH, and Prince HM: Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol Ther 2013;21:2122–2129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Heczey A, Louis CU, Savoldo B, Dakhova O, Durett A, Grilley B, Liu H, Wu MF, Mei Z, Gee A, Mehta B, Zhang H, Mahmood N, Tashiro H, Heslop HE, Dotti G, Rooney CM, and Brenner MK: CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther 2017;25:2214–2224 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Yvon E, Del Vecchio M, Savoldo B, Hoyos V, Dutour A, Anichini A, Dotti G, and Brenner MK: Immunotherapy of metastatic melanoma using genetically engineered GD2-specific T cells. Clin Cancer Res 2009;15:5852–5860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Prapa M, Caldrer S, Spano C, Bestagno M, Golinelli G, Grisendi G, Petrachi T, Conte P, Horwitz EM, Campana D, Paolucci P, and Dominici M: A novel anti-GD2/4-1BB chimeric antigen receptor triggers neuroblastoma cell killing. Oncotarget 2015;6:24884–24894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Yankelevich M, Kondadasula SV, Thakur A, Buck S, Cheung NK, and Lum LG: Anti-CD3 × anti-GD2 bispecific antibody redirects T-cell cytolytic activity to neuroblastoma targets. Pediatr Blood Cancer 2012;59:1198–1205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Cheng M, Santich BH, Xu H, Ahmed M, Huse M, and Cheung NK: Successful engineering of a highly potent single-chain variable-fragment (scFv) bispecific antibody to target disialoganglioside (GD2) positive tumors. Oncoimmunology 2016;5:e1168557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Rashidijahanabad Z, and Huang X: Recent advances in tumor associated carbohydrate antigen based chimeric antigen receptor T cells and bispecific antibodies for anti-cancer immunotherapy. Semin Immunol 2020;47:101390. [DOI] [PMC free article] [PubMed] [Google Scholar]