Crimson Publishers Publish With Us Reprints e-Books Video articles

Full Text

Gastroenterology Medicine & Research

Current Advances in Immunotherapy for Colorectal Cancer

Li Ma1,2,3, Stefan Moisyadi1,2 and Wen-Ming Chu1,4,5*

1Manoa Institute for Life Science and Cancer, Honolulu, Hawaii 96813, USA

2Institute for Biogenesis Research, University of Hawaii, Honolulu, Hawaii 96822, USA

3MBBE Program, Graduate School, University of Hawaii, Honolulu, Hawaii 96822, USA

4Sbarro Institute, Philadelphia, Pennsylvania 19122, USA

5Department of Quantitative Health Sciences, University of Hawaii, Honolulu, Hawaii 96813,USA

*Corresponding author:Wen-Ming Chu, Manoa Institute for Life Science and Cancer, Honolulu, Hawaii 96813, USA

Submission:June 26, 2024;Published: July 05, 2024

DOI: 10.31031/GMR.2023.07.000675

ISSN 2637-7632
Volume7 Issue5

Abstract

Purpose: This study aimed to briefly review the current advances in immunotherapy for colorectal cancer.
Methods: We conducted a brief review of the latest literature.
Results: We discuss the advantages and challenges of variable forms of immunotherapy for colorectal cancer
Conclusion: Immunotherapy holds great promise for colorectal cancer.

Keywords:mRNA; Immunotherapy; PD-1; PD-L1; Monoclonal antibody (mAb); Nanobody (Nb); CAR-T; CAR-NK; Colorectal cancer; TME

Introduction

Colorectal Cancer (CRC) is the third most common cancer in the United States1. Although the incidence of CRC has been reduced in people over 55 years old, it has markedly increased in those under 55 years old [1]. In the early-stages, CRC usually shows no symptoms and is difficult to identify. Once it is diagnosed, the majority of CRC have already become metastatic CRC (mCRC) leading it to being the second leading cause of cancer-associated death in the United States [1]. Therefore, there is an urgent need for better therapeutic strategies for the disease. Cancer immunotherapy has transformed CRC treatment by harnessing the innate capabilities of the host immune system to combat and eliminate cancer. Cancer immunotherapy has come in a variety of forms including immune checkpoint inhibitors, cancer vaccination and immune cell therapy.

Immune checkpoint inhibitors are monoclonal antibodies (mAbs) targeting programmed death-1 (PD-1), PD-1 ligand 1 (PD-L1), and cytotoxic T-lymphocyte antigen 4 (CTLA-4). The PD-1 mAb has been approved by the Food and Drug Administration (FDA) as the first-line treatment and anti-PD-L1 mAb as the second-line treatment for mCRC. However, only 5% of mCRC patients respond to anti-PD-1 therapy. These mCRC patients harbor mutations in mismatch repair genes leading to high microsatellite instability (MSI-H) and the accumulation of somatic mutations. The somatic mutations are found in about 15% of CRC patients causing the expression of neoantigens. These neoantigens are perceived as foreign by the body triggering adaptive immune responses and subsequent tumor infiltration of innate immune cells and T cells and their expression of immune checkpoint molecules in the Tumor Microenvironment (TME) of “hot CRC”. Conversely, approximately 85% of CRC cases exhibit low microsatellite instability (MSI-L) or Microsatellite (MSS), characterized by the absence of tumor-infiltrating innate immune cells and T cells, as well asl the lack of immune checkpoint expression on these cells in the TME of “cold CRC”. Moreover, there are abundant Myeloid Derived Suppressor Cells (MDSCs) and Tumor-Associate Macrophages (TAMs) in the TME of cold CRC, impeding the effectiveness of anti-PD-1 and anti-PD-L1 therapy. Furthermore, mAbs are large molecules and have limited tumor penetration capacity diminishing therapeutic efficacy of anti-PD-1 and anti-PD-L1 therapy for mCRC.

Nanobodies (Nbs) and single-chain variable fragments (scFvs) derived from IgG of monoclonal antibodies targeting immune checkpoints are small molecules characterized by low immunogenicity and the ability to efficiently penetrate tumor tissues [2-4]. They are excellent alternatives for treating CRCs. However, these Nbs and scFvs have a short serum half-life hindering their anti-CRC immunity capabilities. A strategy of fusing a Nb with Fc fragment of IgG that increases the size and slows down the renal clearance rate has been successfully used to generate a recombinant PD-L1Nb-Fc (KN035-Fc), which has been approved to treat MSI-H solid tumors including mCRC with frequent administrations [5,6]. The cell immunotherapy via modified T cells, chimeric antigen receptor-T (CAR-T) cells and CAR-natural killer (CAR-NK) cells has been used to treat CRC in pre-clinical and clinical trials. Typically, patients’ own T cells or NK cells are transduced by lentiviral particles containing CAR DNA constructs which selectively target and eliminate tumors. Research findings indicated that a patient with MSI-H mCRC, who developed resistance to the PD-1 inhibitor pembrolizumab, achieved stable disease survival for six months following treatment with T cell receptor-(TCR)-redirected T cells targeting mutant transforming growth factor beta receptor 2 [7]. Pre-clinical and clinical trials using a CAR-T or a CAR-NK strategy to treat mCRC have recently been reported or reviewed elsewhere [8,9]. However, while engineered CAR-T cells have cured patients with untreatable hematological cancers, the CAR-T therapy faces challenges with poor cell penetration into solid tumors, cytokine release syndrome (CRS), ‘on-target, off tumor toxicity’ and genotoxicity [10].

Messenger RNA technology has emerged as a powerful platform that enhances the effectiveness of immunotherapy. Lipid Nanoparticles (LNPs) encapsulating In Vitro Transcribed (IVT), nucleoside-modified (N1-methylpseudouridine-containing) mRNA vaccines against COVID-19 have proven to be a promising and highly successful approach in mitigating the severe impacts of the COVID-19 pandemic. This IVT mRNA-LNP technology has rapidly integrated into immunotherapy forming the platform of mRNA vaccines, mRNA-driven production of mAbs, Nbs and proteins of interest, and mRNA-associated CAR-T and CAR-NK cell therapy. This platform provides standardized procedures within well-equipped facilities, efficiently delivering mRNA into cells to avoid genotoxicity and reduce immunogenicity and potential Cytokine Release Syndrome (CRS). It also enables rapid and continuous production of monoclonal antibodies, nanobodies, and proteins either in cells or in vivo [10-12]. For example, once a tumor associated protein or a specific antigen is identified from tumors, an mRNA construct is quickly engineered and then is used either to transfect Dendritic Cells (DCs) producing DC vaccines or to be delivered directly to the host, triggering adaptive immunity targeting the tumor [13- 17]. Moreover, rapid CAR-mRNA constructs can replace CAR-DNA constructs based on nanobody- or/and bispecific antibodies-scFvs accelerating utility and effectiveness of CAR-T or CAR-NK strategy [8,18-20]. We have recently adopted this strategy to produce anti- PD-L1 Nbs in mice in a xenograft mouse CRC model. We found that LNP-encapsulated PD-L1Nb-mRNA significantly suppressed CRC progression (Chu et al. unpublished data).

Future Prospective

CRC tumorigenesis is a consequence of multi-faceted effects, and a single-agent therapy is unlikely to eliminate CRC tumors. Moreover, there is an unmet need for strategies that precisely deliver components of immunotherapy to the tumor site and modify the Tumor Microenvironment (TME) in cold CRC. These strategies are expected to enhance effectiveness and reduce adverse effects. Furthermore, integrated immunotherapeutic approaches utilizing mRNA platforms, nanobody/scFvs, synthetic Notch/specific receptors, probiotic-guided CAR-T/NK cells, immune checkpoint inhibitors, vaccines, and innate immune agonists will be more powerful and efficacious in the treatment of solid tumors, including CRC [12,13,17,19,21].

References

  1. Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A (2023) Colorectal cancer statistics, 2023. CA Cancer J Clin 73(3): 233-254.
  2. Salvador JP, Vilaplana L, Marco MP (2019) Nanobody: Outstanding features for diagnostic and therapeutic applications. Anal Bioanal Chem 411(9): 1703-1713.
  3. Rahbarizadeh F, Ahmadvand D, Sharifzadeh Z (2011) Nanobody; an old concept and new vehicle for immunotargeting. Immunol Invest 40(3): 299-338.
  4. Yu S, Xiong G, Zhao S, Tang Y, Tang H, et al. (2021) Nanobodies targeting immune checkpoint molecules for tumor immunotherapy and immunoimaging (Review). Int J Mol Med 47(2): 444-454.
  5. Yang W, Zheng H, Lv W, Zhu Y (2023) Current status and prospect of immunotherapy for colorectal cancer. Int J Colorectal Dis 38(1): 266.
  6. Zhang F (2017) Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov 3: 17004.
  7. Maggadottir SM (2024) Transient TCR-based T cell therapy in a patient with advanced treatment-resistant MSI-high colorectal cancer. Molecular Therapy 32(6): 2021-2029.
  8. Kamrani A (2024) New immunotherapy approaches for colorectal cancer: focusing on CAR-T cell, BiTE, and oncolytic viruses. Cell Commun Sig 22(1): 56.
  9. Xiao L, et al. (2019) Adoptive transfer of NKG2D CAR mRNA-engineered natural killer cells in colorectal cancer patients. Mol Ther 27(6): 1114-1125.
  10. Baker DJ, Arany Z, Baur JA, Epstein JA, June CH (2023) CAR T therapy beyond cancer: The evolution of a living drug. Nature 619(7971): 707-715.
  11. Pardi N, Hogan MJ, Porter FW, Weissman D (2018) mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov 17(4): 261-279.
  12. Deal CE, Carfi A, Plante OJ (2021) Advancements in mRNA encoded antibodies for passive immunotherapy. Vaccines (Basel) 9(2): 108.
  13. Zong Y, Lin Y, Wei T, Cheng Q (2023) Lipid Nanoparticle (LNP) enables mRNA delivery for cancer therapy. Adv Mater 35(51): e230326.
  14. Verbeke R, Hogan MJ, Lore K, Pardi N (2022) Innate immune mechanisms of mRNA vaccines. Immunity 55(11): 1993-2005.
  15. Lorentzen CL, Haanen JB, Met O, Svane IM (2022) Clinical advances and ongoing trials on mRNA vaccines for cancer treatment. Lancet Oncol 23(10): e450-e458.
  16. Palmer CD, Rappaport AR, Davis MJ, Hart MG, Scallan CD, et al. (2022) Individualized, heterologous chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced metastatic solid tumors: phase 1 trial interim results. Nat Med 28(8): 1619-1629.
  17. Ben-Akiva E, Karlsson J, Hemmati S, Yu H, Tzeng SY, et al. (2023) Biodegradable lipophilic polymeric mRNA nanoparticles for ligand-free targeting of splenic dendritic cells for cancer vaccination. Proc Natl Acad Sci U S A 120(26): e2301606120.
  18. Mo F, Duan S, Jiang X, Yang X, Hou X, et al. (2021) Nanobody-based chimeric antigen receptor T cells designed by CRISPR/Cas9 technology for solid tumor immunotherapy. Signal Transduct Target Ther 6: 80.
  19. Vincent RL, Gurbatri CR, Li F, Vardoshvili A, Coker C, et al. (2023) Probiotic-guided CAR-T cells for solid tumor targeting. Science 382(6667): 211-218.
  20. Taheri FH, Hassani M, Sharifzadeh Z, Behdani M, Abdoli S, et al. (2024) Tuning spacer length improves the functionality of the nanobody-based VEGFR2 CAR T cell. BMC Biotechnol 24(1): 1.
  21. Roybal KT, Williams JZ, Morsut L, Rupp LJ, Kolinko I, et al. (2016) Engineering T cells with customized therapeutic response programs using synthetic notch receptors. Cell 167(2): 419-432 e416.

© 2024 Wen-Ming Chu, This is an open access article distributed under the terms of the Creative Commons Attribution License , which permits unrestricted use, distribution, and build upon your work non-commercially.

About Crimson

We at Crimson Publishing are a group of people with a combined passion for science and research, who wants to bring to the world a unified platform where all scientific know-how is available read more...

Leave a comment

Contact Info

  • Crimson Publishers, LLC
  • 260 Madison Ave, 8th Floor
  •     New York, NY 10016, USA
  • +1 (929) 600-8049
  • +1 (929) 447-1137
  • info@crimsonpublishers.com
  • www.crimsonpublishers.com