Next Article in Journal
Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update
Next Article in Special Issue
Real-World Experience of Pembrolizumab Monotherapy in Patients with Recurrent or Persistent Cervical Cancer: A Korean Multi-Center Retrospective Study (KGOG1041)
Previous Article in Journal
Effect of 1α,25(OH)2 Vitamin D3 in Mutant P53 Glioblastoma Cells: Involvement of Neutral Sphingomyelinase1
Previous Article in Special Issue
The Oncoprotein SKI Acts as A Suppressor of NK Cell-Mediated Immunosurveillance in PDAC
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Challenges and Opportunities for Effective Cancer Immunotherapies

by
Clare Y. Slaney
1,2,* and
Michael H. Kershaw
1,2,*
1
Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia
2
Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3000, Australia
*
Authors to whom correspondence should be addressed.
Cancers 2020, 12(11), 3164; https://doi.org/10.3390/cancers12113164
Submission received: 19 October 2020 / Accepted: 24 October 2020 / Published: 28 October 2020
(This article belongs to the Special Issue Challenges and Opportunities for Effective Cancer Immunotherapies)
Using immunotherapy to treat cancers can be traced back to the 1890s, where a New York physician William Coley used heat-killed bacteria to treat cancer patients, which became known as “Coley’s toxin”. Of the almost 900 cancer patients he treated, some tumours regressed and some patients were free from recurrence for a number of years [1]. However, the toxin component was inconsistent, patients’ reactions were unpredictable and the anti-cancer mechanism was not known. With the advancement of radiation therapy and chemotherapy in the 20th century, Coley’s toxin was not used anymore.
In the past ten years, we have witnessed many revolutionary immunotherapies being approved to use in the clinic for treating cancer patients. These immunotherapies include the first cancer vaccine, Sipuleucel-T for advanced prostate cancer; checkpoint inhibitors such as ipilimumab, pembrolizumab and nivolumab for the treatment of advanced melanoma and other solid cancers; oncolytic virus T-Vec for melanoma; a bispecific cancer-directed T-cell engager, blinatumomab, for the treatment of acute lymphoblastic leukemia, and chimeric antigen receptor (CAR) T cells for treating certain lymphoma and leukemias [2,3]. Together, these immunotherapies have had a remarkable impact on clinical outcomes.
This year, although the majority of the world is locked down in response to the coronavirus disease (COVID-19) pandemic, a number of cancer immunotherapies were approved by the US Food and Drug Administration (FDA). The newly approved treatments include atezolizumab for advanced melanoma, brexucabtagene autoleucel (Tecartus) for mantle cell lymphoma, which is the third FDA-approved CAR T-cell therapy, pembrolizumab as the first line of treatment for colorectal cancer and pembrolizumab for cutaneous squamous cell carcinoma.
Although more and more treatment options are becoming available, challenges still remain. Immune checkpoint inhibitors (ICIs) work for certain cancer types such as melanoma, but not all cancer types respond. Even in melanoma, half of the patients do not achieve a significant beneficial response, and a substantial number of responding patients experience cancer relapse after the initial response [4]. Unfortunately, these ICI therapeutics are also often associated with a high rate of toxicity, with severe toxicities occurring in approximately 20–50% of patients [5]. Other immunotherapies can have similar problems. Certain cancers such as pancreatic cancer have proven to be difficult to treat using all the current available immunotherapies [6].
Building on the success of ICIs, numerous immunotherapies have been tested to be used in combination with other immunotherapies or with some already existing treatments. For example, anti-PD1 has been tested in combination with CAR T-cell therapy [7], oncolytic virus treatment [8,9], cyclin-dependent kinase inhibitors [10]. Given the potency of the treatment components as monotherapies, it is not surprising that a number of these combinations led to synergistic efficacy. With an abundance of combination immunotherapy trials ongoing, more and more factors that influence the therapeutic success have been revealed and synergistic design of different combination therapies may provide optimal benefit to the patients with different types of cancers.
Although yet to demonstrate efficacy in solid tumours, enormous efforts have been made in CAR T-cell research. These include the discovery of new tumour antigen targets [11], more options for combination therapy [7,12], creating T-cell products with a more desirable phenotype [13], improved manufacturing protocols [14], and novel methods for enhancing in vivo expansion of the CAR T cells [15,16,17]. Although most of the current immunotherapies have focused on T cells, other cellular therapies such as those utilising NK cell cytotoxicity [18,19], dendritic cells [20] and macrophages [21] are also under investigation.
Another extensively explored area lies in the understanding of immunosuppression of the tumour microenvironment (TME) [22]. The TME consists of tumour cells, immune cells, stroma, extracellular matrix and some soluble factors. This complex environment plays a fundamental role in tumour progression, shapes the tumour immune response and eventually determines the efficacy of immunotherapies [23,24,25]. A number of strategies have been developed in the past few years to shift the TME to favour anti-tumour immunity, and clinical studies have validated several biomarkers of the TME predicting tumour responsiveness to immunotherapies [26,27].
Much knowledge has accumulated in the past ten years, and the cancer immunotherapy field is moving forward at a fast pace. Many current obstacles will likely be overcome through improved knowledge, more advances in treatment technologies [28] and the identification of new cancer targets. In addition, new combination treatments incorporating immunotherapies, and the identification of predictive biomarkers for cancer immunotherapies, may lead to further effective treatments utilizing the immune system for a wide range of cancers.

Funding

The authors are supported by grants from the National Health and Medical Research Council and the National Breast Cancer Foundation (NBCF) of Australia.

Acknowledgments

We are grateful for all the contributing authors for their contribution to this Special Issue and the support from the Cancers editorial staff.

Conflicts of Interest

The authors declare no potential conflicts of interest.

References

  1. Coley, W.B. The Treatment of Malignant Tumors by Repeated Inoculations of Erysipelas. With a report of ten original cases; 1893. Available online: https://pubmed.ncbi.nlm.nih.gov/1984929/ (accessed on 19 October 2020).
  2. Slaney, C.Y.; Wang, P.; Darcy, P.K.; Kershaw, M.H. CARs versus BiTEs: A Comparison between T Cell-Redirection Strategies for Cancer Treatment. Cancer Discov. 2018, 8, 924–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kershaw, M.H.; Westwood, J.A.; Darcy, P.K. Gene-engineered T cells for cancer therapy. Nat. Rev. Cancer 2013, 13, 525–541. [Google Scholar] [CrossRef] [PubMed]
  4. Testori, A.A.E.; Chiellino, S.; van Akkooi, A.C.J. Adjuvant Therapy for Melanoma: Past, Current, and Future Developments. Cancers 2020, 12, 1994. [Google Scholar] [CrossRef]
  5. Johnson, D.B.; Reynolds, K.L.; Sullivan, R.J.; Balko, J.M.; Patrinely, J.R.; Cappelli, L.C.; Naidoo, J.; Moslehi, J.J. Immune checkpoint inhibitor toxicities: Systems-based approaches to improve patient care and research. Lancet Oncol. 2020, 21, e398–e404. [Google Scholar] [CrossRef] [PubMed]
  6. Ali, A.I.; Oliver, A.J.; Samiei, T.; Chan, J.D.; Kershaw, M.H.; Slaney, C.Y. Genetic Redirection of T Cells for the Treatment of Pancreatic Cancer. Front. Oncol. 2019, 9, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. John, L.B.; Devaud, C.; Duong, C.P.; Yong, C.S.; Beavis, P.A.; Haynes, N.M.; Chow, M.T.; Smyth, M.J.; Kershaw, M.H.; Darcy, P.K. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 2013, 19, 5636–5646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2018, 174, 1031–1032. [Google Scholar] [CrossRef] [PubMed]
  9. Long, G.V.; Dummer, R.; Ribas, A.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.S.; et al. Efficacy analysis of MASTERKEY-265 phase 1b study of talimogene laherparepvec (T-VEC) and pembrolizumab (pembro) for unresectable stage IIIB-IV melanoma. J. Clin. Oncol. 2016, 34, 9568. [Google Scholar] [CrossRef]
  10. Hossain, D.M.S.; Javaid, S.; Cai, M.; Zhang, C.; Sawant, A.; Hinton, M.; Sathe, M.; Grein, J.; Blumenschein, W.; Pinheiro, E.M.; et al. Dinaciclib induces immunogenic cell death and enhances anti-PD1-mediated tumor suppression. J. Clin. Invest. 2018, 128, 644–654. [Google Scholar] [CrossRef]
  11. Apavaloaei, A.; Hardy, M.P.; Thibault, P.; Perreault, C. The Origin and Immune Recognition of Tumor-Specific Antigens. Cancers 2020, 12, 2607. [Google Scholar] [CrossRef]
  12. Mardiana, S.; John, L.B.; Henderson, M.A.; Slaney, C.Y.; von Scheidt, B.; Giuffrida, L.; Davenport, A.J.; Trapani, J.A.; Neeson, P.J.; Loi, S.; et al. A multifunctional role for adjuvant anti-4-1BB therapy in augmenting anti-tumor response by chimeric antigen receptor T cells. Cancer Res. 2017. [Google Scholar] [CrossRef] [Green Version]
  13. Cieri, N.; Camisa, B.; Cocchiarella, F.; Forcato, M.; Oliveira, G.; Provasi, E.; Bondanza, A.; Bordignon, C.; Peccatori, J.; Ciceri, F.; et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2013, 121, 573–584. [Google Scholar] [CrossRef]
  14. Wang, X.; Popplewell, L.L.; Wagner, J.R.; Naranjo, A.; Blanchard, M.S.; Mott, M.R.; Norris, A.P.; Wong, C.W.; Urak, R.Z.; Chang, W.C.; et al. Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL. Blood 2016, 127, 2980–2990. [Google Scholar] [CrossRef]
  15. Chan, J.D.; von Scheidt, B.; Zeng, B.; Oliver, A.J.; Davey, A.S.; Ali, A.I.; Thomas, R.; Trapani, J.A.; Darcy, P.K.; Kershaw, M.H.; et al. Enhancing chimeric antigen receptor T-cell immunotherapy against cancer using a nanoemulsion-based vaccine targeting cross-presenting dendritic cells. Clin. Transl. Immunol. 2020, 9, e1157. [Google Scholar] [CrossRef]
  16. Von Scheidt, B.; Wang, M.; Oliver, A.J.; Chan, J.D.; Jana, M.K.; Ali, A.I.; Clow, F.; Fraser, J.D.; Quinn, K.M.; Darcy, P.K.; et al. Enterotoxins can support CAR T cells against solid tumors. Proc. Natl. Acad. Sci. USA 2019, 116, 25229–25235. [Google Scholar] [CrossRef]
  17. Slaney, C.Y.; von Scheidt, B.; Davenport, A.J.; Beavis, P.; Westwood, J.A.; Mardiana, S.; Tscharke, D.; Ellis, S.; Prince, H.M.; Trapani, J.A.; et al. Dual-specific chimeric antigen receptor T cells and an indirect vaccine eradicate a variety of large solid tumors in an immunocompetent, self-antigen setting. Clin. Cancer Res. 2017. [Google Scholar] [CrossRef] [Green Version]
  18. Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. EBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef] [PubMed]
  19. Ponath, V.; Frech, M.; Bittermann, M.; Al Khayer, R.; Neubauer, A.; Brendel, C.; Pogge von Strandmann, E. The Oncoprotein SKI Acts as A Suppressor of NK Cell-Mediated Immunosurveillance in PDAC. Cancers 2020, 12, 2857. [Google Scholar] [CrossRef]
  20. Bol, K.F.; Schreibelt, G.; Rabold, K.; Wculek, S.K.; Schwarze, J.K.; Dzionek, A.; Teijeira, A.; Kandalaft, L.E.; Romero, P.; Coukos, G.; et al. The clinical application of cancer immunotherapy based on naturally circulating dendritic cells. J. Immunother Cancer 2019, 7, 109. [Google Scholar] [CrossRef]
  21. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E.; et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef]
  22. Beavis, P.A.; Slaney, C.Y.; Kershaw, M.H.; Neeson, P.J.; Darcy, P.K. Enhancing the efficacy of adoptive cellular therapy by targeting tumor-induced immunosuppression. Immunotherapy 2015, 7, 499–512. [Google Scholar] [CrossRef]
  23. Oliver, A.J.; Darcy, P.K.; Kershaw, M.H.; Slaney, C.Y. Tissue-specific tumour microenvironments are an emerging determinant of immunotherapy responses. J. Thorac. Dis. 2020, 12, 4504–4509. [Google Scholar] [CrossRef] [PubMed]
  24. Oliver, A.J.; Davey, A.S.; Keam, S.P.; Mardiana, S.; Chan, J.D.; von Scheidt, B.; Beavis, P.A.; House, I.G.; Van Audernaerde, J.R.; Darcy, P.K.; et al. Tissue-specific tumor microenvironments influence responses to immunotherapies. Clin. Transl. Immunol. 2019, 8, e1094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Hass, R. Role of MSC in the Tumor Microenvironment. Cancers 2020, 12, 2107. [Google Scholar] [CrossRef]
  26. Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef] [PubMed]
  27. Oliver, A.J.; Lau, P.K.H.; Unsworth, A.S.; Loi, S.; Darcy, P.K.; Kershaw, M.H.; Slaney, C.Y. Tissue-Dependent Tumor Microenvironments and Their Impact on Immunotherapy Responses. Front. Immunol. 2018, 9, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Maruoka, Y.; Furusawa, A.; Okada, R.; Inagaki, F.; Wakiyama, H.; Kato, T.; Nagaya, T.; Choyke, P.L.; Kobayashi, H. Interleukin-15 after Near-Infrared Photoimmunotherapy (NIR-PIT) Enhances T Cell Response against Syngeneic Mouse Tumors. Cancers 2020, 12, 2575. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Slaney, C.Y.; Kershaw, M.H. Challenges and Opportunities for Effective Cancer Immunotherapies. Cancers 2020, 12, 3164. https://doi.org/10.3390/cancers12113164

AMA Style

Slaney CY, Kershaw MH. Challenges and Opportunities for Effective Cancer Immunotherapies. Cancers. 2020; 12(11):3164. https://doi.org/10.3390/cancers12113164

Chicago/Turabian Style

Slaney, Clare Y., and Michael H. Kershaw. 2020. "Challenges and Opportunities for Effective Cancer Immunotherapies" Cancers 12, no. 11: 3164. https://doi.org/10.3390/cancers12113164

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