1
|
Olivares-Hernández R, Riaño-Umbarila L, Becerril B, Alagón A, Vázquez-López H. Pharmacokinetic evaluation of a single chain antibody fragment against scorpion toxins in sheep. Toxicon 2024; 242:107691. [PMID: 38522587 DOI: 10.1016/j.toxicon.2024.107691] [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: 10/29/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
A key aspect during the development of antivenoms is the evaluation of the efficiency and security of the therapeutic molecules. In this work, we report the pharmacokinetic analysis of a neutralizing single chain antibody fragment named LR (scFv LR) where three sheep were used as a large animal model. The animals were injected through i.v. route with 2 mg of scFv LR. Blood samples were drawn every minute within the first 15 min, the sampling continues at 20, 25, 30, 45, 60, 90, 120 min, subsequently at 1-h intervals, 3, 4, 5, 6 h, two more samples at 9 and 12 h and, two more samples at 24 and 48 h and finally at one-day intervals during 4 days. scFv LR levels were measured from blood serum and urine samples by an ELISA. The pharmacokinetics of the experimental data was analyzed using the three-exponential kinetics. The value of the fast initial component (τ1=0.409±0.258min) indicated that the scFv is distributed rapidly into the tissues. The mean residence time, MRT, was 45 ± 0.51 min and the clearance (CL), 114.3 ± 14.3 mL/min. From urine samples it was possible to detect significant amounts of scFv LR, which is evidence of renal elimination.
Collapse
Affiliation(s)
- Roberto Olivares-Hernández
- Departamento de Procesos y Tecnología, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Cuajimalpa de Morelos, Ciudad de México, 05348, Mexico
| | - Lidia Riaño-Umbarila
- Investigadora por México, CONAHCYT. Instituto de Biotecnología-Universidad Nacional Autónoma de México, Mexico; Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología-Universidad Nacional Autónoma de México. Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos, 62210, Mexico
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología-Universidad Nacional Autónoma de México. Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos, 62210, Mexico
| | - Alejandro Alagón
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología-Universidad Nacional Autónoma de México. Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos, 62210, Mexico
| | - Hilda Vázquez-López
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología-Universidad Nacional Autónoma de México. Avenida Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos, 62210, Mexico.
| |
Collapse
|
2
|
Mathias-Machado MC, de Jesus VHF, Jácome A, Donadio MD, Aruquipa MPS, Fogacci J, Cunha RG, da Silva LM, Peixoto RD. Claudin 18.2 as a New Biomarker in Gastric Cancer-What Should We Know? Cancers (Basel) 2024; 16:679. [PMID: 38339430 PMCID: PMC10854563 DOI: 10.3390/cancers16030679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Gastric cancer (GC) remains a formidable global health challenge, ranking among the top-five causes of cancer-related deaths worldwide. The majority of patients face advanced stages at diagnosis, with a mere 6% five-year survival rate. First-line treatment for metastatic GC typically involves a fluoropyrimidine and platinum agent combination; yet, predictive molecular markers have proven elusive. This review navigates the evolving landscape of GC biomarkers, with a specific focus on Claudin 18.2 (CLDN18.2) as an emerging and promising target. Recent phase III trials have unveiled the efficacy of Zolbetuximab, a CLDN18.2-targeting antibody, in combination with oxaliplatin-based chemotherapy for CLDN18.2-positive metastatic GC. As this novel therapeutic avenue unfolds, understanding the nuanced decision making regarding the selection of anti-CLDN18.2 therapies over other targeted agents in metastatic GC becomes crucial. This manuscript reviews the evolving role of CLDN18.2 as a biomarker in GC and explores the current status of CLDN18.2-targeting agents in clinical development. The aim is to provide concise insights into the potential of CLDN18.2 as a therapeutic target and guide future clinical decisions in the management of metastatic GC.
Collapse
Affiliation(s)
- Maria Cecília Mathias-Machado
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, São Paulo 04538-132, Brazil; (M.D.D.); (M.P.S.A.); (R.D.P.)
| | | | - Alexandre Jácome
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, Belo Horizonte 30360-680, Brazil;
| | - Mauro Daniel Donadio
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, São Paulo 04538-132, Brazil; (M.D.D.); (M.P.S.A.); (R.D.P.)
| | | | - João Fogacci
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, Rio de Janeiro 22775-003, Brazil;
| | - Renato Guerino Cunha
- Cellular Therapy Program, Division of Hematology, Oncoclínicas, São Paulo 04538-132, Brazil;
| | | | - Renata D’Alpino Peixoto
- Division of Gastrointestinal Medical Oncology, Oncoclínicas, São Paulo 04538-132, Brazil; (M.D.D.); (M.P.S.A.); (R.D.P.)
| |
Collapse
|
3
|
Schriek AI, Aldon YLT, van Gils MJ, de Taeye SW. Next-generation bNAbs for HIV-1 cure strategies. Antiviral Res 2024; 222:105788. [PMID: 38158130 DOI: 10.1016/j.antiviral.2023.105788] [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: 10/10/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Despite the ability to suppress viral replication using anti-retroviral therapy (ART), HIV-1 remains a global public health problem. Curative strategies for HIV-1 have to target and eradicate latently infected cells across the body, i.e. the viral reservoir. Broadly neutralizing antibodies (bNAbs) targeting the HIV-1 envelope glycoprotein (Env) have the capacity to neutralize virions and bind to infected cells to initiate elimination of these cells. To improve the efficacy of bNAbs in terms of viral suppression and viral reservoir eradication, next generation antibodies (Abs) are being developed that address the current limitations of Ab treatment efficacy; (1) low antigen (Env) density on (reactivated) HIV-1 infected cells, (2) high viral genetic diversity, (3) exhaustion of immune cells and (4) short half-life of Abs. In this review we summarize and discuss preclinical and clinical studies in which anti-HIV-1 Abs demonstrated potent viral control, and describe the development of engineered Abs that could address the limitations described above. Next generation Abs with optimized effector function, avidity, effector cell recruitment and immune cell activation have the potential to contribute to an HIV-1 cure or durable control.
Collapse
Affiliation(s)
- A I Schriek
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands.
| | - Y L T Aldon
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - M J van Gils
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - S W de Taeye
- Amsterdam UMC Location University of Amsterdam, Department of Medical Microbiology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Mehrizi TZ, Mirzaei M, Ardestani MS. Pegylation, a Successful Strategy to Address the Storage and Instability Problems of Blood Products: Review 2011-2021. Curr Pharm Biotechnol 2024; 25:247-267. [PMID: 37218184 DOI: 10.2174/1389201024666230522091958] [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: 08/02/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 05/24/2023]
Abstract
Conjugation of polyethylene glycol (PEGylation) to blood proteins and cells has emerged as a successful approach to address some of the issues attributed to the storage of blood products, including their short half-life and instability. In this regard, this review study aims to compare the influence of different PEGylation strategies on the quality of several blood products like red blood cells (RBCs), platelets, plasma proteins, i.e., albumin, coagulation factor VIII, and antibodies. The results indicated that conjugating succinimidyl carbonate methoxyPEG (SCmPEG) to platelets could improve blood transfusion safety by preventing these cells from being attached to low-load hidden bacteria in blood products. Moreover, coating of 20 kD succinimidyl valerate (SVA)-mPEG to RBCs was able to extend the half-life and stability of these cells during storage, as well as immune camouflage their surface antigens to prevent alloimmunisation. As regards albumin products, PEGylation improved the albumin stability, especially during sterilization, and there was a relationship between the molecular weight (MW) of PEG molecules and the biological half-life of the conjugate. Although coating antibodies with short-chain PEG molecules could enhance their stabilities, these modified proteins were cleared from the blood faster. Also, branched PEG molecules enhanced the retention and shielding of the fragmented and bispecific antibodies. Overall, the results of this literature review indicate that PEGylation can be considered a useful tool for enhancing the stability and storage of blood components.
Collapse
Affiliation(s)
| | - Mehdi Mirzaei
- Iran Ministry of Health and Medical Education, Deputy Ministry for Education, Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Zhang C, D'Angelo D, Buttini F, Yang M. Long-acting inhaled medicines: Present and future. Adv Drug Deliv Rev 2024; 204:115146. [PMID: 38040120 DOI: 10.1016/j.addr.2023.115146] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Inhaled medicines continue to be an essential part of treatment for respiratory diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis. In addition, inhalation technology, which is an active area of research and innovation to deliver medications via the lung to the bloodstream, offers potential advantages such as rapid onset of action, enhanced bioavailability, and reduced side effects for local treatments. Certain inhaled macromolecules and particles can also end up in different organs via lymphatic transport from the respiratory epithelium. While the majority of research on inhaled medicines is focused on the delivery technology, particle engineering, combination therapies, innovations in inhaler devices, and digital health technologies, researchers are also exploring new pharmaceutical technologies and strategies to prolong the duration of action of inhaled drugs. This is because, in contrast to most inhaled medicines that exert a rapid onset and short duration of action, long-acting inhaled medicines (LAIM) improve not only the patient compliance by reducing the dosing frequency, but also the effectiveness and convenience of inhaled therapies to better manage patients' conditions. This paper reviews the advances in LAIM, the pharmaceutical technologies and strategies for developing LAIM, and emerging new inhaled modalities that possess a long-acting nature and potential in the treatment and prevention of various diseases. The challenges in the development of the future LAIM are also discussed where active research and innovations are taking place.
Collapse
Affiliation(s)
- Chengqian Zhang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Davide D'Angelo
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Francesca Buttini
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Mingshi Yang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016, Shenyang, China.
| |
Collapse
|
6
|
Schengrund CL. The Ying and Yang of Ganglioside Function in Cancer. Cancers (Basel) 2023; 15:5362. [PMID: 38001622 PMCID: PMC10670608 DOI: 10.3390/cancers15225362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
The plethora of information about the expression of cancer cell-associated gangliosides, their role(s) in signal transduction, and their potential usefulness in the development of cancer treatments makes this an appropriate time to review these enigmatic glycosphingolipids. Evidence, reflecting the work of many, indicates that (1) expression of specific gangliosides, not generally found in high concentrations in most normal human cells, can be linked to certain types of cancer. (2) Gangliosides can affect the ability of cells to interact either directly or indirectly with growth factor receptors, thereby changing such things as a cell's mobility, rate of proliferation, and metastatic ability. (3) Anti-ganglioside antibodies have been tested, with some success, as potential treatments for certain cancers. (4) Cancer-associated gangliosides shed into the circulation can (a) affect immune cell responsiveness either positively or negatively, (b) be considered as diagnostic markers, and (c) be used to look for recurrence. (5) Cancer registries enable investigators to evaluate data from sufficient numbers of patients to obtain information about potential therapies. Despite advances that have been made, a discussion of possible approaches to identifying additional treatment strategies to inhibit metastasis, responsible for the majority of deaths of cancer patients, as well as for treating therapy-resistant tumors, is included.
Collapse
Affiliation(s)
- Cara-Lynne Schengrund
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
7
|
Kalinovsky DV, Kholodenko IV, Svirshchevskaya EV, Kibardin AV, Ryazantsev DY, Rozov FN, Larin SS, Deyev SM, Kholodenko RV. Targeting GD2-Positive Tumor Cells by Pegylated scFv Fragment-Drug Conjugates Carrying Maytansinoids DM1 and DM4. Curr Issues Mol Biol 2023; 45:8112-8125. [PMID: 37886955 PMCID: PMC10604934 DOI: 10.3390/cimb45100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Oligomerization of antibody fragments via modification with polyethylene glycol (pegylation) may alter their function and properties, leading to a multivalent interaction of the resulting constructs with the target antigen. In a recent study, we generated pegylated monomers and multimers of scFv fragments of GD2-specific antibodies using maleimide-thiol chemistry. Multimerization enhanced the antigen-binding properties and demonstrated a more efficient tumor uptake in a syngeneic GD2-positive mouse cancer model compared to monomeric antibody fragments, thereby providing a rationale for improving the therapeutic characteristics of GD2-specific antibody fragments. In this work, we obtained pegylated conjugates of scFv fragments of GD2-specific antibodies with maytansinoids DM1 or DM4 using tetravalent PEG-maleimide (PEG4). The protein products from the two-stage thiol-maleimide reaction resolved by gel electrophoresis indicated that pegylated scFv fragments constituted the predominant part of the protein bands, and most of the scFv formed pegylated monomers and dimers. The conjugates retained the ability to bind ganglioside GD2 comparable to that of the parental scFv fragment and to specifically interact with GD2-positive cells. Both induced significant inhibitory effects in the GD2-positive B78-D14 cell line, in contrast to the GD2-negative B16 cell line. The decrease in the B78-D14 cell viability when treated with scFv-PEG4-DM4 was more prominent than that for scFv-PEG4-DM1, and was characterized by a twofold lower half-maximal inhibitory concentration (IC50). Unlike the parental scFv fragment, the product of scFv and PEG4 conjugation (scFv-PEG4), consisting predominantly of pegylated scFv multimers and monomers, induced direct cell death in the GD2-positive B78-D14 cells. However, the potency of scFv-PEG4 was low in the selected concentration range, thus demonstrating that the cytotoxic effect of DM1 and DM4 within the antibody fragment-drug conjugates was primary. The suggested approach may contribute to development of novel configurations of antibody fragment-drug conjugates for cancer treatment.
Collapse
Affiliation(s)
- Daniel V. Kalinovsky
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., Moscow 119121, Russia
| | - Elena V. Svirshchevskaya
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Alexey V. Kibardin
- Laboratory of Molecular Immunology, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia; (A.V.K.); (S.S.L.)
| | - Dmitry Yu. Ryazantsev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Fedor N. Rozov
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
| | - Sergey S. Larin
- Laboratory of Molecular Immunology, D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., Moscow 117997, Russia; (A.V.K.); (S.S.L.)
| | - Sergey M. Deyev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
- Laboratory of Molecular Pharmacology, Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., Moscow 119992, Russia
- “Biomarker” Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan 420008, Russia
| | - Roman V. Kholodenko
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho-Maklaya St., Moscow 117997, Russia; (D.V.K.); (E.V.S.); (D.Y.R.); (F.N.R.); (S.M.D.)
- Real Target LLC, Miklukho-Maklaya St., 16/10, Moscow 117997, Russia
| |
Collapse
|
8
|
Khilji SK, Op 't Hoog C, Warschkau D, Lühle J, Goerdeler F, Freitag A, Seeberger PH, Moscovitz O. Smaller size packs a stronger punch - Recent advances in small antibody fragments targeting tumour-associated carbohydrate antigens. Theranostics 2023; 13:3041-3063. [PMID: 37284439 PMCID: PMC10240822 DOI: 10.7150/thno.80901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/26/2023] [Indexed: 06/08/2023] Open
Abstract
Attached to proteins, lipids, or forming long, complex chains, glycans represent the most versatile post-translational modification in nature and surround all human cells. Unique glycan structures are monitored by the immune system and differentiate self from non-self and healthy from malignant cells. Aberrant glycosylations, termed tumour-associated carbohydrate antigens (TACAs), are a hallmark of cancer and are correlated with all aspects of cancer biology. Therefore, TACAs represent attractive targets for monoclonal antibodies for cancer diagnosis and therapy. However, due to the thick and dense glycocalyx as well as the tumour micro-environment, conventional antibodies often suffer from restricted access and limited effectiveness in vivo. To overcome this issue, many small antibody fragments have come forth, showing similar affinity with better efficiency than their full-length counterparts. Here we review small antibody fragments against specific glycans on tumour cells and highlight their advantages over conventional antibodies.
Collapse
Affiliation(s)
- Sana Khan Khilji
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Charlotte Op 't Hoog
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Graduate School of Life Sciences, Utrecht University, 3584 CH Utrecht, Netherlands
| | - David Warschkau
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jost Lühle
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Felix Goerdeler
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Anika Freitag
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Peter H. Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Oren Moscovitz
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| |
Collapse
|
9
|
Mistareehi A, Bendowski KT, Bizanti A, Madas J, Zhang Y, Kwiat AM, Nguyen D, Kogut N, Ma J, Chen J, Cheng ZJ. Topographical distribution and morphology of SP-IR axons in the antrum, pylorus, and duodenum of mice. Auton Neurosci 2023; 246:103074. [PMID: 36804650 PMCID: PMC10515648 DOI: 10.1016/j.autneu.2023.103074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/30/2023]
Abstract
Substance-P (SP) is a commonly used marker of nociceptive afferent axons, and it plays an important role in a variety of physiological functions including the regulation of motility, gut secretion, and vascular flow. Previously, we found that SP-immunoreactive (SP-IR) axons densely innervated the pyloric antrum of the flat-mount of the mouse whole stomach muscular layer. However, the regional distribution and morphology of SP-IR axons in the submucosa and mucosa were not well documented. In this study, the mouse antrum-pylorus-duodenum (APD) were transversely and longitudinally sectioned. A Zeiss M2 imager was used to scan the serial sections of each APD (each section montage consisted of 50-100 all-in-focus maximal projection images). To determine the detailed structures of SP-IR axons and terminals, we used the confocal microscope to scan the regions of interest. We found that 1) SP-IR axons innervated the muscular, submucosal, and mucosal layers. 2) In the muscular layer, SP-IR varicose axons densely innervated the muscles and formed varicose terminals which encircled myenteric neurons. 3) In the submucosa, SP-IR axons innervated blood vessels and submucosal ganglia and formed a network in Brunner's glands. 4) In the mucosa, SP-IR axons innervated the muscularis mucosae. Some SP-IR axons entered the lamina propria. 5) The muscular layer of the antrum and duodenum showed a higher SP-IR axon density than the pyloric sphincter. 6) SP-IR axons were from extrinsic and intrinsic origins. This work provided a comprehensive view of the distribution and morphology of SP-IR axons in the APD at single cell/axon/varicosity scale. This data will be used to create a 3D scaffold of the SP-IR axon innervation of the APD.
Collapse
Affiliation(s)
- Anas Mistareehi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Kohlton T Bendowski
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Ariege Bizanti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Jazune Madas
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Yuanyuan Zhang
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Andrew M Kwiat
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Duyen Nguyen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Nicole Kogut
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Jichao Ma
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, United States of America.
| |
Collapse
|
10
|
Huysamen A, Fadeyi OE, Mayuni G, Dogbey DM, Mungra N, Biteghe FAN, Hardcastle N, Ramamurthy D, Akinrinmade OA, Naran K, Cooper S, Lang D, Richter W, Hunter R, Barth S. Click Chemistry-Generated Auristatin F-Linker-Benzylguanine for a SNAP-Tag-Based Recombinant Antibody-Drug Conjugate Demonstrating Selective Cytotoxicity toward EGFR-Overexpressing Tumor Cells. ACS OMEGA 2023; 8:4026-4037. [PMID: 36743041 PMCID: PMC9893251 DOI: 10.1021/acsomega.2c06844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/03/2023] [Indexed: 06/18/2023]
Abstract
Antibody-drug conjugates (ADCs) are bifunctional molecules combining the targeting potential of monoclonal antibodies with the cancer-killing ability of cytotoxic drugs. This simple yet intelligently designed system directly addresses the lack of specificity encountered with conventional anti-cancer treatment regimes. However, despite their initial success, the generation of clinically sustainable and effective ADCs has been plagued by poor tumor penetration, undefined chemical linkages, unpredictable pharmacokinetic profiles, and heterogeneous mixtures of products. To this end, we generated a SNAP-tag-based fusion protein targeting the epidermal growth factor receptor (EGFR)-a biomarker of aggressive and drug-resistant cancers. Here, we demonstrate the use of a novel click coupling strategy to engineer a benzylguanine (BG)-linker-auristatin F (AuriF) piece that can be covalently tethered to the EGFR-targeting SNAP-tag-based fusion protein in an irreversible 1:1 stoichiometric reaction to form a homogeneous product. Furthermore, using these recombinant ADCs to target EGFR-overexpressing tumor cells, we provide a proof-of-principle for generating biologically active antimitotic therapeutic proteins capable of inducing cell death in a dose-dependent manner, thus alleviating some of the challenges of early ADC development.
Collapse
Affiliation(s)
- Allan
M. Huysamen
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Olaolu E. Fadeyi
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Grace Mayuni
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dennis M. Dogbey
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Neelakshi Mungra
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Centre
for Immunity and Immunotherapies, Seattle
Children’s Research Institute, Seattle, Washington 98101, United States
| | - Fleury A. N. Biteghe
- Department
of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, California 90048, United States
| | - Natasha Hardcastle
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dharanidharan Ramamurthy
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Olusiji A. Akinrinmade
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Department
of Molecular Pharmacology, Albert Einstein
College of Medicine, Bronx, New York 10461, United States
| | - Krupa Naran
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Susan Cooper
- Division
of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | - Dirk Lang
- Division
of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | | | - Roger Hunter
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Stefan Barth
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- South
African Research Chair in Cancer Biotechnology, Department of Integrative
Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape
Town 7700, South Africa
| |
Collapse
|
11
|
Minibody-Based and scFv-Based Antibody Fragment-Drug Conjugates Selectively Eliminate GD2-Positive Tumor Cells. Int J Mol Sci 2023; 24:ijms24021239. [PMID: 36674755 PMCID: PMC9860947 DOI: 10.3390/ijms24021239] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Ganglioside GD2 is a well-established target expressed on multiple solid tumors, many of which are characterized by low treatment efficiency. Antibody-drug conjugates (ADCs) have demonstrated marked success in a number of solid tumors, and GD2-directed drug conjugates may also hold strong therapeutic potential. In a recent study, we showed that ADCs based on the approved antibody dinutuximab and the drugs monomethyl auristatin E (MMAE) or F (MMAF) manifested potent and selective cytotoxicity in a panel of tumor cell lines and strongly inhibited solid tumor growth in GD2-positive mouse cancer models. Here, we employed two different GD2-binding moieties-minibodies and scFv fragments that carry variable antibody domains identical to those of dinutuximab, and site-directly conjugated them to MMAE or MMAF by thiol-maleimide chemistry with drug-to-antibody ratios (DAR) of 2 and 1, respectively. Specific binding of the antibody fragment-drug conjugates (FDCs) to GD2 was confirmed in direct ELISA, flow cytometry, and confocal microscopy. Selective cytotoxic and cytostatic effects of the conjugates were observed in GD2-positive but not GD2-negative neuroblastoma and melanoma cell lines. Minibody-based FDCs demonstrated more pronounced cytotoxic effects and stronger antigen binding compared to scFv-based FDCs. The developed molecules may offer considerable practical benefit, since antibody fragment-drug conjugates are capable of enhancing therapeutic efficacy of ADCs by improving their pharmacokinetic characteristics and reducing side effects.
Collapse
|
12
|
Lou H, Cao X. Antibody variable region engineering for improving cancer immunotherapy. Cancer Commun (Lond) 2022; 42:804-827. [PMID: 35822503 PMCID: PMC9456695 DOI: 10.1002/cac2.12330] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/25/2022] [Accepted: 06/22/2022] [Indexed: 04/09/2023] Open
Abstract
The efficacy and specificity of conventional monoclonal antibody (mAb) drugs in the clinic require further improvement. Currently, the development and application of novel antibody formats for improving cancer immunotherapy have attracted much attention. Variable region-retaining antibody fragments, such as antigen-binding fragment (Fab), single-chain variable fragment (scFv), bispecific antibody, and bi/trispecific cell engagers, are engineered with humanization, multivalent antibody construction, affinity optimization and antibody masking for targeting tumor cells and killer cells to improve antibody-based therapy potency, efficacy and specificity. In this review, we summarize the application of antibody variable region engineering and discuss the future direction of antibody engineering for improving cancer therapies.
Collapse
Affiliation(s)
- Hantao Lou
- Ludwig Institute of Cancer ResearchUniversity of OxfordOxfordOX3 7DRUK
- Chinese Academy for Medical Sciences Oxford InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
| | - Xuetao Cao
- Chinese Academy for Medical Sciences Oxford InstituteNuffield Department of MedicineUniversity of OxfordOxfordOX3 7FZUK
- Department of ImmunologyCentre for Immunotherapy, Institute of Basic Medical SciencesChinese Academy of Medical SciencesBeijing100005P. R. China
| |
Collapse
|
13
|
Kim YS, Potashnikova DM, Gisina AM, Kholodenko IV, Kopylov AT, Tikhonova OV, Kurbatov LK, Saidova AA, Tvorogova AV, Kholodenko RV, Belousov PV, Vorobjev IA, Zgoda VG, Yarygin KN, Lupatov AY. TRIM28 Is a Novel Regulator of CD133 Expression Associated with Cancer Stem Cell Phenotype. Int J Mol Sci 2022; 23:9874. [PMID: 36077272 PMCID: PMC9456468 DOI: 10.3390/ijms23179874] [Citation(s) in RCA: 4] [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: 07/18/2022] [Revised: 08/24/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
CD133 is an extensively studied marker of the most malignant tumor cell population, designated as cancer stem cells (CSCs). However, the function of this glycoprotein and its involvement in cell regulatory cascades are still poorly understood. Here we show a positive correlation between the level of CD133 plasma membrane expression and the proliferative activity of cells of the Caco-2, HT-29, and HUH7 cancer cell lines. Despite a substantial difference in the proliferative activities of cell populations with different levels of CD133 expression, transcriptomic and proteomic profiling revealed only minor distinctions between them. Nonetheless, a further in silico assessment of the differentially expressed transcripts and proteins revealed 16 proteins that could be involved in the regulation of CD133 expression; these were assigned ranks reflecting the apparent extent of their involvement. Among them, the TRIM28 transcription factor had the highest rank. The prominent role of TRIM28 in CD133 expression modulation was confirmed experimentally in the Caco2 cell line clones: the knockout, though not the knockdown, of the TRIM28 gene downregulated CD133. These results for the first time highlight an important role of the TRIM28 transcription factor in the regulation of CD133-associated cancer cell heterogeneity.
Collapse
Affiliation(s)
- Yan S. Kim
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Daria M. Potashnikova
- Cell Biology and Histology Department, School of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Alisa M. Gisina
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Arthur T. Kopylov
- Laboratory of Systems Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Olga V. Tikhonova
- Laboratory of Systems Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Leonid K. Kurbatov
- Transcriptome Analysis Group, Analytical Branch Department, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Aleena A. Saidova
- Cell Biology and Histology Department, School of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
- Department of Transcription Factors, V.A. Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V. Tvorogova
- Laboratory of Cell Motility, A.N. Belozersky Research Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Roman V. Kholodenko
- Laboratory of Molecular Immunology, M.M. Shemyakin–Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Pavel V. Belousov
- Endocrinology Research Centre, 117292 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, V.A. Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ivan A. Vorobjev
- Laboratory of Cell Motility, A.N. Belozersky Research Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
- Laboratory of Biophotonics and Imaging, National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
| | - Victor G. Zgoda
- Laboratory of Systems Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Alexey Yu. Lupatov
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| |
Collapse
|
14
|
Kalinovsky DV, Kibardin AV, Kholodenko IV, Svirshchevskaya EV, Doronin II, Konovalova MV, Grechikhina MV, Rozov FN, Larin SS, Deyev SM, Kholodenko RV. Therapeutic efficacy of antibody-drug conjugates targeting GD2-positive tumors. J Immunother Cancer 2022; 10:e004646. [PMID: 35764367 PMCID: PMC9240879 DOI: 10.1136/jitc-2022-004646] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Both ganglioside GD2-targeted immunotherapy and antibody-drug conjugates (ADCs) have demonstrated clinical success as solid tumor therapies in recent years, yet no research has been carried out to develop anti-GD2 ADCs against solid tumors. This is the first study to analyze cytotoxic activity of clinically relevant anti-GD2 ADCs in a wide panel of cell lines with varying GD2 expression and their effects in mouse models of GD2-positive solid cancer. METHODS Anti-GD2 ADCs were generated based on the GD2-specific antibody ch14.18 approved for the treatment of neuroblastoma and commonly used drugs monomethyl auristatin E (MMAE) or F (MMAF), conjugated via a cleavable linker by thiol-maleimide chemistry. The antibody was produced in a mammalian expression system, and its specific binding to GD2 was analyzed. Antigen-binding properties and biodistribution of the ADCs in mice were studied in comparison with the parent antibody. Cytotoxic effects of the ADCs were evaluated in a wide panel of GD2-positive and GD2-negative tumor cell lines of neuroblastoma, glioma, sarcoma, melanoma, and breast cancer. Their antitumor effects were studied in the B78-D14 melanoma and EL-4 lymphoma syngeneic mouse models. RESULTS The ch14.18-MMAE and ch14.18-MMAF ADCs retained antigen-binding properties of the parent antibody. Direct dependence of the cytotoxic effect on the level of GD2 expression was observed in cell lines of different origin for both ADCs, with IC50 below 1 nM for the cells with high GD2 expression and no cytotoxic effect for GD2-negative cells. Within the analyzed cell lines, ch14.18-MMAF was more effective in the cells overexpressing GD2, while ch14.18-MMAE had more prominent activity in the cells expressing low GD2 levels. The ADCs had a similar biodistribution profile in the B78-D14 melanoma model compared with the parent antibody, reaching 7.7% ID/g in the tumor at 48 hours postinjection. The average tumor size in groups treated with ch14.18-MMAE or ch14.18-MMAF was 2.6 times and 3.8 times smaller, respectively, compared with the control group. Antitumor effects of the anti-GD2 ADCs were also confirmed in the EL-4 lymphoma model. CONCLUSION These findings validate the potential of ADCs targeting ganglioside GD2 in treating multiple GD2-expressing solid tumors.
Collapse
Affiliation(s)
- Daniel V Kalinovsky
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexey V Kibardin
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Moscow, Russia
| | | | - Elena V Svirshchevskaya
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Igor I Doronin
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Real Target LLC, Moscow, Russia
| | - Mariya V Konovalova
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Maria V Grechikhina
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey S Larin
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Moscow, Russia
| | - Sergey M Deyev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Roman V Kholodenko
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Real Target LLC, Moscow, Russia
| |
Collapse
|
15
|
Cao W, Xing H, Li Y, Tian W, Song Y, Jiang Z, Yu J. Claudin18.2 is a novel molecular biomarker for tumor-targeted immunotherapy. Biomark Res 2022; 10:38. [PMID: 35642043 PMCID: PMC9153115 DOI: 10.1186/s40364-022-00385-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/16/2022] [Indexed: 12/18/2022] Open
Abstract
The claudin18.2 (CLDN18.2) protein, an isoform of claudin18, a member of the tight junction protein family, is a highly selective biomarker with limited expression in normal tissues and often abnormal expression during the occurrence and development of various primary malignant tumors, such as gastric cancer/gastroesophageal junction (GC/GEJ) cancer, breast cancer, colon cancer, liver cancer, head and neck cancer, bronchial cancer and non-small-cell lung cancer. CLDN18.2 participates in the proliferation, differentiation and migration of tumor cells. Recent studies have identified CLDN18.2 expression as a potential specific marker for the diagnosis and treatment of these tumors. With its specific expression pattern, CLDN18.2 has become a unique molecule for targeted therapy in different cancers, especially in GC; for example, agents such as zolbetuximab (claudiximab, IMAB362), a monoclonal antibody (mAb) against CLDN18.2, have been developed. In this review, we outline recent advances in the development of immunotherapy strategies targeting CLDN18.2, including monoclonal antibodies (mAbs), bispecific antibodies (BsAbs), chimeric antigen receptor T (CAR-T) cells redirected to target CLDN18.2, and antibody–drug conjugates (ADCs).
Collapse
Affiliation(s)
- Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Haizhou Xing
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wenliang Tian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
16
|
Denizci Öncü M, Balcıoğlu BK, Özgür B, Öztürk HÜ, Serhatlı M, Işık Ş, Erdağ B, Dinler Doğanay G, Özdemir Bahadır A. Structure-based engineering of an antiangiogenic scFv antibody for soluble production in E. coli without loss of activity. Biotechnol Appl Biochem 2021; 69:2122-2137. [PMID: 34694021 DOI: 10.1002/bab.2273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/18/2021] [Indexed: 11/09/2022]
Abstract
Development of monoclonal antibody therapeutics against vascular endothelial growth factor receptor 2 (VEGFR-2) protein, which is the main regulator in angiogenesis, has been a major challenge for years. In the current study, we engineer an inclusion body forming single-chain variable fragment (scFv) against VEGFR-2 by using complementarity determining regions (CDR) grafting technique to improve its solubility and investigate the activity of the engineered molecule. CDR sequences of the target scFv were grafted into the framework of another intrinsically soluble scFv molecule. Based on the computational results, CDR grafting has increased the solubility of the grafted scFv molecule. Results confirmed that the grafting approach increased in vivo folding properties of the target scFv molecule compared with the original scFv molecule. Similar binding affinities to the VEGFR-2 were observed for the original and the grafted scFv by surface plasmon resonance assays. Biological activity assays, including human umbilical vein endothelial cells proliferation and wound healing assays, showed that grafted scFv molecule has an antiangiogenic property. This study suggests that an antiangiogenic scFv fully expressed as an inclusion body can be rescued by grafting its CDR regions to a scFv expressed in a soluble form without any loss in its binding property and its activity.
Collapse
Affiliation(s)
- Melis Denizci Öncü
- Genetic Engineering and Biotechnology Institute, Marmara Research Center, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey.,Molecular Biology and Genetics Department, İstanbul Technical University, Istanbul, Turkey
| | - Bertan Koray Balcıoğlu
- Genetic Engineering and Biotechnology Institute, Marmara Research Center, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey
| | - Beytullah Özgür
- Genetic Engineering and Biotechnology Institute, Marmara Research Center, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey
| | - Hasan Ümit Öztürk
- Genetic Engineering and Biotechnology Institute, Marmara Research Center, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey
| | - Müge Serhatlı
- Genetic Engineering and Biotechnology Institute, Marmara Research Center, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey
| | - Şeyma Işık
- Medical Biotechnology Department, Acıbadem University, Istanbul, Turkey
| | - Berrin Erdağ
- Health Sciences Department, İstanbul Aydın University, Istanbul, Turkey
| | - Gizem Dinler Doğanay
- Molecular Biology and Genetics Department, İstanbul Technical University, Istanbul, Turkey
| | - Aylin Özdemir Bahadır
- Genetic Engineering and Biotechnology Institute, Marmara Research Center, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey
| |
Collapse
|
17
|
Kholodenko IV, Kim YS, Gisina AM, Lupatov AY, Kholodenko RV, Yarygin KN. Analysis of the Correlation between CD133 Expression on Human Colorectal Adenocarcinoma Cells HT-29 and Their Resistance to Chemotherapeutic Drugs. Bull Exp Biol Med 2021; 171:156-163. [PMID: 34057619 DOI: 10.1007/s10517-021-05188-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 12/25/2022]
Abstract
A correlation was found between chemoresistance of HT-29CD133+ and HT-29CD133- sublines obtained after cell sorting and high expression of CD133. On the other hand, knockout of the PROM1 gene and, as a consequence, the absence of CD133 expression did not increase the sensitivity of tumor cells to chemotherapy, which indicates the absence of a direct effect of CD133 on the formation of chemoresistance in colorectal cancer cells. Variants of the HT-29 line with complete or partial knockout of the PROM1 gene were equally sensitive to protein kinase inhibitors sorafenib and sunitinib. Notably, the highest resistance to mTOR inhibitors, temsirolimus and everolimus, was shown by cells with complete knockout of the PROM1 gene (KO-HT-29 (P1)). These findings suggest that CD133 is associated with the chemoresistance of colorectal cancer cells, but is not involved in its formation.
Collapse
Affiliation(s)
- I V Kholodenko
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia.
| | - Ya S Kim
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - A M Gisina
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - A Yu Lupatov
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - R V Kholodenko
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - K N Yarygin
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
18
|
Schengrund CL. Gangliosides and Neuroblastomas. Int J Mol Sci 2020; 21:E5313. [PMID: 32726962 PMCID: PMC7432824 DOI: 10.3390/ijms21155313] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/09/2020] [Accepted: 07/18/2020] [Indexed: 12/19/2022] Open
Abstract
The focus of this review is the ganglio-series of glycosphingolipids found in neuroblastoma (NB) and the myriad of unanswered questions associated with their possible role(s) in this cancer. NB is one of the more common solid malignancies of children. Five-year survival for those diagnosed with low risk NB is 90-95%, while that for children with high-risk NB is around 40-50%. Much of the survival rate reflects age of diagnosis with children under a year having a much better prognosis than those over two. Identification of expression of GD2 on the surface of most NB cells led to studies of the effectiveness and subsequent approval of anti-GD2 antibodies as a treatment modality. Despite much success, a subset of patients, possibly those whose tumors fail to express concentrations of gangliosides such as GD1b and GT1b found in tumors from patients with a good prognosis, have tumors refractory to treatment. These observations support discussion of what is known about control of ganglioside synthesis, and their actual functions in NB, as well as their possible relationship to treatment response.
Collapse
Affiliation(s)
- Cara-Lynne Schengrund
- Department of Biochemistry and Molecular Biology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
19
|
Nazha B, Inal C, Owonikoko TK. Disialoganglioside GD2 Expression in Solid Tumors and Role as a Target for Cancer Therapy. Front Oncol 2020; 10:1000. [PMID: 32733795 PMCID: PMC7358363 DOI: 10.3389/fonc.2020.01000] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Gangliosides are carbohydrate-containing sphingolipids that are widely expressed in normal tissues, making most subtypes unsuitable as targets for cancer therapy. However, the disialoganglioside GD2 subtype has limited expression in normal tissues but is overexpressed across a wide range of tumors. Disialoganglioside GD2 can be considered a tumor-associated antigen and well-suited as a target for cancer therapy. Disialoganglioside GD2 is implicated in tumor development and malignant phenotypes through enhanced cell proliferation, motility, migration, adhesion, and invasion, depending on the tumor type. This provides a rationale for targeting disialoganglioside GD2 in cancer therapy with the development of anti-GD2 monoclonal antibodies and other therapeutic approaches. Anti-GD2 monoclonal antibodies target GD2-expressing tumor cells, leading to phagocytosis and destruction by means of antibody-dependent cell-mediated cytotoxicity, lysis by complement-dependent cytotoxicity, and apoptosis and necrosis through direct induction of cell death. Anti-GD2 monoclonal antibodies may also prevent homing and adhesion of circulating malignant cells to the extracellular matrix. Disialoganglioside GD2 is highly expressed by almost all neuroblastomas, by most melanomas and retinoblastomas, and by many Ewing sarcomas and, to a more variable degree, by small cell lung cancer, gliomas, osteosarcomas, and soft tissue sarcomas. Successful treatment of disialoganglioside GD2-expressing tumors with anti-GD2 monoclonal antibodies is hindered by pharmacologic factors such as insufficient antibody affinity to mediate antibody-dependent cell-mediated cytotoxicity, inadequate penetration of antibody into the tumor microenvironment, and toxicity related to disialoganglioside GD2 expression by normal tissues such as peripheral sensory nerve fibers. Nonetheless, anti-GD2 monoclonal antibody dinutuximab (ch14.18) has been approved by the U.S. Food and Drug Administration and dinutuximab beta (ch14.18/CHO) has been approved by the European Medicines Agency for the treatment of high-risk neuroblastoma in pediatric patients. Clinical trials of anti-GD2 therapy are currently ongoing in patients with other types of disialoganglioside GD2-expressing tumors as well as neuroblastoma. In addition to anti-GD2 monoclonal antibodies, anti-GD2 therapeutic approaches include chimeric antigen receptor T-cell therapy, disialoganglioside GD2 vaccines, immunocytokines, immunotoxins, antibody-drug conjugates, radiolabeled antibodies, targeted nanoparticles, and T-cell engaging bispecific antibodies. Clinical trials should clarify further the potential of anti-GD2 therapy for disialoganglioside GD2-expressing malignant tumors.
Collapse
Affiliation(s)
- Bassel Nazha
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| | - Cengiz Inal
- Salem Veterans Affairs Medical Center, Salem, VA, United States
| | - Taofeek K. Owonikoko
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
20
|
Sorokin M, Kholodenko I, Kalinovsky D, Shamanskaya T, Doronin I, Konovalov D, Mironov A, Kuzmin D, Nikitin D, Deyev S, Buzdin A, Kholodenko R. RNA Sequencing-Based Identification of Ganglioside GD2-Positive Cancer Phenotype. Biomedicines 2020; 8:E142. [PMID: 32486168 PMCID: PMC7344710 DOI: 10.3390/biomedicines8060142] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
The tumor-associated ganglioside GD2 represents an attractive target for cancer immunotherapy. GD2-positive tumors are more responsive to such targeted therapy, and new methods are needed for the screening of GD2 molecular tumor phenotypes. In this work, we built a gene expression-based binary classifier predicting the GD2-positive tumor phenotypes. To this end, we compared RNA sequencing data from human tumor biopsy material from experimental samples and public databases as well as from GD2-positive and GD2-negative cancer cell lines, for expression levels of genes encoding enzymes involved in ganglioside biosynthesis. We identified a 2-gene expression signature combining ganglioside synthase genes ST8SIA1 and B4GALNT1 that serves as a more efficient predictor of GD2-positive phenotype (Matthews Correlation Coefficient (MCC) 0.32, 0.88, and 0.98 in three independent comparisons) compared to the individual ganglioside biosynthesis genes (MCC 0.02-0.32, 0.1-0.75, and 0.04-1 for the same independent comparisons). No individual gene showed a higher MCC score than the expression signature MCC score in two or more comparisons. Our diagnostic approach can hopefully be applied for pan-cancer prediction of GD2 phenotypes using gene expression data.
Collapse
Affiliation(s)
- Maxim Sorokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., 119992 Moscow, Russia
- Omicsway Corp., 340 S Lemon Ave, 6040, Walnut, CA 91789, USA
| | - Irina Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya St., 119121 Moscow, Russia;
| | - Daniel Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
| | - Tatyana Shamanskaya
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., 117997 Moscow, Russia; (T.S.); (D.K.)
| | - Igor Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Real Target LLC, 108841 Moscow, Russia
| | - Dmitry Konovalov
- D. Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, 1, Samory Mashela St., 117997 Moscow, Russia; (T.S.); (D.K.)
| | - Aleksei Mironov
- Skolkovo Institute of Science and Technology, 3, Nobelya St., 121205 Moscow, Russia;
| | - Denis Kuzmin
- Moscow Institute of Physics and Technology (National Research University), 141700 Moscow, Russia;
| | - Daniil Nikitin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
| | - Sergey Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., 119992 Moscow, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Sechenov First Moscow State Medical University, 8-2, Trubetskaya St., 119992 Moscow, Russia
- Moscow Institute of Physics and Technology (National Research University), 141700 Moscow, Russia;
- Oncobox ltd., 121205 Moscow, Russia
| | - Roman Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10, Miklukho- Maklaya St., 117997 Moscow, Russia; (M.S.); (D.K.); (I.D.); (D.N.); (S.D.); (A.B.)
- Real Target LLC, 108841 Moscow, Russia
| |
Collapse
|
21
|
Effective Strategies to Overcome the Insolubility of Recombinant ScFv Antibody against EpCAM Extracellular Domain in E. coli. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10044-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|