Next Article in Journal
Quantitative ctDNA Detection in Hepatoblastoma: Implications for Precision Medicine
Previous Article in Journal
The β-Secretase 1 Enzyme as a Novel Therapeutic Target for Prostate Cancer
Previous Article in Special Issue
Phase 4 Multinational Multicenter Retrospective and Prospective Real-World Study of Nivolumab in Recurrent and Metastatic Squamous Cell Carcinoma of the Head and Neck
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Targeted Immunotherapies for Cancers

by
Anthony Cheung
1,2,* and
Alicia Chenoweth
1,2
1
Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
2
St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(1), 11; https://doi.org/10.3390/cancers16010011
Submission received: 27 November 2023 / Accepted: 12 December 2023 / Published: 19 December 2023
(This article belongs to the Special Issue Targeted Immunotherapies for Cancers)
Advancements in immunotherapy have revolutionized cancer treatment in a broad variety of hematological and solid malignancies and rejuvenated the field of cancer immunology. Several types of immunotherapies, including adoptive cell transfer and immune checkpoint inhibitors, have achieved durable clinical responses. This Special Issue highlights promising immune targeting strategies that may provide the basis for further translational investigations.
Nivolumab is an anti-PD-1 monoclonal antibody (mAb) which prolonged survival and retained the quality of life in a randomized, open-label phase III trial for patients with recurrent/metastatic squamous cell carcinoma of the head and neck (R/M SCCHN) [1]. It is now approved for use in the US, EU, and Canada for R/M SCCHN patients with progressive disease at or within six months after platinum-based therapy. Gogate et al. conducted a multinational retrospective study to capture the real-world utilization of nivolumab, demonstrating the effectiveness and safety of nivolumab in most of the patients and reporting stability or improvement in their health-related quality of life.
Despite the success in immunogenic tumors, due to the substantial heterogeneity of immune target expression and immune cell infiltration in tumor lesions, only subsets of cancer patients currently benefit from immunotherapy treatments [2,3]. This highlights an urgent unmet need for new immune targets and modification of mAbs for improved clinical function. Novel preclinical studies and clinical trials are investigating drugs targeting the immune checkpoint axis, either as monotherapy or in combination with other regimens [4]. Clinical results of immunotherapy in low immunogenic cancer types are poor, with response rates as low as 5% with monotherapy, especially in unselected patient populations [5,6]. A combination of chemotherapy or targeted therapy, together with checkpoint blockade, could demonstrate better success. Small-molecule drugs targeting CDK2 at efficacious doses may be associated with toxicity [7]. To address this limitation, Cheung et al. reported that in aggressive triple-negative breast cancers (TNBCs), CDK inhibition at suboptimal doses promotes immune cell recruitment to tumors and PD-L1 expression by surviving TNBC cells, and can complement checkpoint inhibitor immunotherapy. In orthotopic TNBC-bearing xenograft mice, suboptimal CDK inhibitor doses given sequentially ahead of dosing with the anti-PD-L1 antibody avelumab significantly restricted tumor growth compared with monotherapy [8].
Sato et al. reported another promising immune targeting strategy via adoptive cell therapy using chimeric antigen receptor engineered T cell (CAR-T) targeting carcinoembryonic antigen (CEA), a key target antigen which is highly expressed on the surface of pancreatic ductal adenocarcinoma (PDAC) cells [9]. They demonstrated that anti-CEA-CAR-T adoptive cell treatment induced regression of CEA-positive PDAC tumors in orthotopic mouse models, and that the CEA expression level was correlated with tumor heterogeneity and could be clinically used as a biomarker to select patients for anti-CEA-CAR-T therapy. Moreover, Christodoulou et al. gathered current data from the literature regarding the clinical testing and limitation of CAR-based therapies against difficult-to-treat acute myeloid leukemia. The authors discussed the potential benefits of using safer alternative lymphocytes, such as CAR-NK cells (natural killer cells with CARs) which has shown promise in preclinical studies [10,11].
Furthermore, a recombinant single-chain fragment variable (scFv) that contains the complete antigen-binding domains of a whole antibody has several advantages, such as stability and specificity against tumor antigens, with proven ability to penetrate tumor tissues and diffuse [12]. Muñoz-López et al. discussed the principle, generation, and applications of scFvs, particularly in the diagnosis and selective treatment of cancer. Breakthroughs in clinical trials have demonstrated that scFvs are safe molecules and present great antitumor activity when incorporated into CAR-T or bispecific T-cell engager (BiTE) systems.
Narbona et al. reported a new optimized version of colorectal cancer-targeted immunotoxin with low immunogenicity and enhanced antitumor activity in solid tumors. The immunogenic nature of bacterial and plant toxins represents a major drawback to their clinical use, as they can be recognized as foreign by the patient’s immune system and lead to the formation of anti-drug antibodies (ADAs) which neutralize and clear the immunotoxin, and can cause immune-related toxicities [13,14]. The authors designed and characterized two different variants of non-immunogenic immunotoxins based on a de-immunized variant of the ribotoxin α-sarcin, one lacking CD4+ T-cell epitopes and one included a recognition and cleavage furin linker to improve the toxin release to the cytosol. The data showed efficient antitumor effects both in vitro and in vivo.
In summary, this Special Issue of Cancers is a collection of articles discussing several immune targeting strategies, including the combination of cell cycle inhibitors in harmony with checkpoint inhibitor immunotherapy, chimeric antigen receptor engineered T or NK cell therapies, scFvs, and optimized immunotoxins. These may provide the basis for further translational investigations, particularly for patients who do not adequately benefit from currently available therapies.
In total, six papers were accepted for publication and inclusion in this Special Issue (comprising four original articles and two reviews).

Conflicts of Interest

The authors declare no conflict of interest.

List of Contributions

  • Gogate, A.; Bennett, B.; Poonja, Z.; Stewart, G.; Medina Colmenero, A.; Szturz, P.; Carrington, C.; Castro, C.; Gemmen, E.; Lau, A.; et al. Phase 4 Multinational Multicenter Retrospective and Prospective Real-World Study of Nivolumab in Recurrent and Metastatic Squamous Cell Carcinoma of the Head and Neck. Cancers 2023, 15, 3552.
  • Cheung, A.; Chenoweth, A.M.; Quist, J.; Sow, H.S.; Malaktou, C.; Ferro, R.; Hoffmann, R.M.; Osborn, G.; Sachouli, E.; French, E.; et al. CDK Inhibition Primes for Anti-PD-L1 Treatment in Triple-Negative Breast Cancer Models. Cancers 2022, 14, 3361.
  • Sato, O.; Tsuchikawa, T.; Kato, T.; Amaishi, Y.; Okamoto, S.; Mineno, J.; Takeuchi, Y.; Sasaki, K.; Nakamura, T.; Umemoto, K.; et al. Tumor Growth Suppression of Pancreatic Cancer Orthotopic Xenograft Model by CEA-Targeting CAR-T Cells. Cancers 2023, 15, 601.
  • Christodoulou, I.; Solomou, E.E. A Panorama of Immune Fighters Armored with CARs in Acute Myeloid Leukemia. Cancers 2023, 15, 3054.
  • Muñoz-López, P.; Ribas-Aparicio, R.M.; Becerra-Báez, E.I.; Fraga-Pérez, K.; Flores-Martínez, L.F.; Mateos-Chávez, A.A.; Luria-Pérez, R. Single-Chain Fragment Variable: Recent Progress in Cancer Diagnosis and Therapy. Cancers 2022, 14, 4206.
  • Narbona, J.; Gordo, R.G.; Tome-Amat, J.; Lacadena, J. A New Optimized Version of a Colorectal Cancer-Targeted Immunotoxin Based on a Non-Immunogenic Variant of the Ribotoxin alpha-Sarcin. Cancers 2023, 15, 1114.

References

  1. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
  2. Pitt, J.M.; Vetizou, M.; Daillere, R.; Roberti, M.P.; Yamazaki, T.; Routy, B.; Lepage, P.; Boneca, I.G.; Chamaillard, M.; Kroemer, G.; et al. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016, 44, 1255–1269. [Google Scholar] [CrossRef] [PubMed]
  3. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef] [PubMed]
  4. Kaplon, H.; Crescioli, S.; Chenoweth, A.; Visweswaraiah, J.; Reichert, J.M. Antibodies to watch in 2023. MAbs 2023, 15, 2153410. [Google Scholar] [CrossRef] [PubMed]
  5. Adams, S.; Schmid, P.; Rugo, H.S.; Winer, E.P.; Loirat, D.; Awada, A.; Cescon, D.W.; Iwata, H.; Campone, M.; Nanda, R.; et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: Cohort A of the phase II KEYNOTE-086 study. Ann. Oncol. 2019, 30, 397–404. [Google Scholar] [CrossRef]
  6. Dirix, L.Y.; Takacs, I.; Jerusalem, G.; Nikolinakos, P.; Arkenau, H.T.; Forero-Torres, A.; Boccia, R.; Lippman, M.E.; Somer, R.; Smakal, M.; et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: A phase 1b JAVELIN Solid Tumor study. Breast Cancer Res. Treat. 2018, 167, 671–686. [Google Scholar] [CrossRef] [PubMed]
  7. Merrick, K.A.; Wohlbold, L.; Zhang, C.; Allen, J.J.; Horiuchi, D.; Huskey, N.E.; Goga, A.; Shokat, K.M.; Fisher, R.P. Switching Cdk2 on or off with small molecules to reveal requirements in human cell proliferation. Mol. Cell 2011, 42, 624–636. [Google Scholar] [CrossRef] [PubMed]
  8. Cheung, A.; Chenoweth, A.M.; Quist, J.; Sow, H.S.; Malaktou, C.; Ferro, R.; Hoffmann, R.M.; Osborn, G.; Sachouli, E.; French, E.; et al. CDK Inhibition Primes for Anti-PD-L1 Treatment in Triple-Negative Breast Cancer Models. Cancers 2022, 14, 3361. [Google Scholar] [CrossRef] [PubMed]
  9. Gansauge, S.; Gansauge, F.; Beger, H.G. Molecular oncology in pancreatic cancer. J. Mol. Med. 1996, 74, 313–320. [Google Scholar] [CrossRef] [PubMed]
  10. Caruso, S.; De Angelis, B.; Del Bufalo, F.; Ciccone, R.; Donsante, S.; Volpe, G.; Manni, S.; Guercio, M.; Pezzella, M.; Iaffaldano, L.; et al. Safe and effective off-the-shelf immunotherapy based on CAR.CD123-NK cells for the treatment of acute myeloid leukaemia. J. Hematol. Oncol. 2022, 15, 163. [Google Scholar] [CrossRef] [PubMed]
  11. Albinger, N.; Pfeifer, R.; Nitsche, M.; Mertlitz, S.; Campe, J.; Stein, K.; Kreyenberg, H.; Schubert, R.; Quadflieg, M.; Schneider, D.; et al. Primary CD33-targeting CAR-NK cells for the treatment of acute myeloid leukemia. Blood Cancer J. 2022, 12, 61. [Google Scholar] [CrossRef] [PubMed]
  12. Munoz-Lopez, P.; Ribas-Aparicio, R.M.; Becerra-Baez, E.I.; Fraga-Perez, K.; Flores-Martinez, L.F.; Mateos-Chavez, A.A.; Luria-Perez, R. Single-Chain Fragment Variable: Recent Progress in Cancer Diagnosis and Therapy. Cancers 2022, 14, 4206. [Google Scholar] [CrossRef] [PubMed]
  13. Sauna, Z.E.; Lagasse, D.; Pedras-Vasconcelos, J.; Golding, B.; Rosenberg, A.S. Evaluating and Mitigating the Immunogenicity of Therapeutic Proteins. Trends Biotechnol. 2018, 36, 1068–1084. [Google Scholar] [CrossRef] [PubMed]
  14. Mazor, R.; King, E.M.; Pastan, I. Strategies to Reduce the Immunogenicity of Recombinant Immunotoxins. Am. J. Pathol. 2018, 188, 1736–1743. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cheung, A.; Chenoweth, A. Targeted Immunotherapies for Cancers. Cancers 2024, 16, 11. https://doi.org/10.3390/cancers16010011

AMA Style

Cheung A, Chenoweth A. Targeted Immunotherapies for Cancers. Cancers. 2024; 16(1):11. https://doi.org/10.3390/cancers16010011

Chicago/Turabian Style

Cheung, Anthony, and Alicia Chenoweth. 2024. "Targeted Immunotherapies for Cancers" Cancers 16, no. 1: 11. https://doi.org/10.3390/cancers16010011

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop