Научная статья на тему 'Immunologic aspects of colorectal cancer progression'

Immunologic aspects of colorectal cancer progression Текст научной статьи по специальности «Клиническая медицина»

CC BY
0
0
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
colorectal cancer / molecular genetic subtypes / tumor-associated macrophages / cytokines / chemokines / колоректальный рак / молекулярно-генетические подтипы / опухоль-ассоциированные макрофаги / цитокины / хемокины

Аннотация научной статьи по клинической медицине, автор научной работы — Anna V. Tishina, Liubov Yu. Vladimirova, Aleksandr B. Sagakyants, Elena A. Dzhenkova, Inna A. Novikova

Colorectal cancer remains in the leading positions in the structures of morbidity and mortality among both sexes. A large number of studies are aimed to reveal new biomarkers targeted at both early diagnosis and improving the effectiveness of drug therapy. Colorectal carcinoma (CC) is heterogeneous in its morphological, molecular and immunological aspects and is a heterogeneous disease. The existing molecular genetic classifications and biomarkers capable of predicting the effectiveness of therapy aren’t optimal enough. New prognostic markers would make it possible to identify a subgroup of patients with a high risk of tumor recurrence, for whom enhanced monitoring and diagnostic monitoring should be established, as well as the selection of highly effective methods in the treatment of colorectal cancer. It has been established that some immune cells in the tumor microenvironment are able to stimulate the development of disease progression. Cytokines and chemokines in the tumor microenvironment stimulate the development of metastases, and their serum levels reflect the current inflammatory response in the tumor tissue. The identification and analysis of immune markers involved in the processes of metastasis and the mechanisms of progression remains an important task of modern medicine. The purpose of the study was to analyze modern ideas about the importance of the immunological microenvironment in the progression of colorectal cancer. The effect of molecular heterogeneity of the tumor on the development of metastases, as well as on resistance to ongoing antitumor therapy. The review reflects the immunological characteristics of CC, including in the context of molecular biological subtypes. It describes the involvement of cells of the immune system (lymphocytes, macrophages) and their products (cytokines, chemokines) in the progression of colorectal cancer, including in the processes of neoangiogenesis, as well as the relationship of the Tand B-cell composition of the tumor microenvironment on the course of the disease. The review also shows the immunogenomic stratification of CC, which can be used to predict the response to immunotherapy for colorectal cancer.

i Надоели баннеры? Вы всегда можете отключить рекламу.

Похожие темы научных работ по клинической медицине , автор научной работы — Anna V. Tishina, Liubov Yu. Vladimirova, Aleksandr B. Sagakyants, Elena A. Dzhenkova, Inna A. Novikova

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Иммунологические аспекты прогрессирования колоректального рака

Колоректальный рак в структурах заболеваемости и смертности среди обоих полов по-прежнему остается на лидирующих позициях. Большое количество исследований нацелено на получение новых биомаркеров, направленных как на раннюю диагностику, так и на улучшение эффективности лекарственной терапии. Колоректальная карцинома неоднородна по своим морфологическим, молекулярным и иммунологическим аспектам и представляет собой гетерогенное заболевание. Существующие молекулярно-генетические классификации и биомаркеры, способные прогнозировать эффективность терапии, неоптимальны. Новые прогностические маркеры позволили бы идентифицировать подгруппу пациентов с высоким риском рецидива опухоли, за которыми должен быть установлен усиленный контроль и диагностическое наблюдение, а также подбор высокоэффективных методов терапии колоректального рака. Установлено, что некоторые иммунные клетки в микроокружении опухоли способны стимулировать развитие прогрессирования заболевания. Цитокины и хемокины в микроокружении опухоли стимулируют развитие метастазов, а их уровни в сыворотке крови отражают текущую воспалительную реакцию в опухолевой ткани. Выявление и анализ иммунных маркеров, участвующих в процессах метастазирования и механизмах прогрессирования, остается важной задачей современной медицины. Целью работы явился анализ современных представлений о значении иммунологического микроокружения, в прогрессировании колоректального рака. Влияние молекулярной гетерогенности опухоли на развитие метастазов, а также на резистентность к проводимой противоопухолевой терапии. В обзоре отражены иммунологические характеристики колоректальной карциномы, в том числе в контексте молекулярнобиологических подтипов. Описывается участие клеток иммунной системы (лимфоцитов, макрофагов) и их продуктов (цитокинов, хемокинов) в прогрессировании колоректального рака, в том числе в процессах неоангиогенеза, а также взаимосвязи Ти В-клеточного состава микроокружения опухоли на течение заболевания. Также в обзоре отображена иммуногеномная стратификация колоректальной карциномы, которая может быть применена для прогнозирования ответа на иммунотерапию колоректального рака.

Текст научной работы на тему «Immunologic aspects of colorectal cancer progression»

South Russian Journal of Cancer. 2024. Vol. 5, No. 1. P. 52-59 https://doi.org/10.37748/2686-9039-2024-5-1-6 https://elibrary.ru/wjwrxu

REVIEW

Immunologic aspects of colorectal cancer progression

A. V. Tishina12, L. Yu. Vladimirova, A. B. Sagakyants, E. A. Dzhenkova, I. A. Novikova, E. Yu. Zlatnik

National Medical Research Centre for Oncology, Rostov-on-Don, Russian Federation

12 tishina@sc-te.ru

Colorectal cancer remains in the leading positions in the structures of morbidity and mortality among both sexes. A large number of studies are aimed to reveal new biomarkers targeted at both early diagnosis and improving the effectiveness of drug therapy. Colorectal carcinoma (CC) is heterogeneous in its morphological, molecular and immunological aspects and is a heterogeneous disease. The existing molecular genetic classifications and biomarkers capable of predicting the effec- tiveness of therapy aren't optimal enough. New prognostic markers would make it possible to identify a subgroup of patients with a high risk of tumor recurrence, for whom enhanced monitoring and diagnostic monitoring should be established, as well as the selection of highly effective methods in the treatment of colorectal cancer. It has been established that some immune cells in the tumor microenvironment are able to stimulate the development of disease progression. Cytokines and chemokines in the tumor microenvironment stimulate the development of metastases, and their serum levels reflect the cur- rent inflammatory response in the tumor tissue. The identification and analysis of immune markers involved in the processes of metastasis and the mechanisms of progression remains an important task of modern medicine. The purpose of the study was to analyze modern ideas about the importance of the immunological microenvironment in the progression of colorectal cancer. The effect of molecular heterogeneity of the tumor on the development of metastases, as well as on resistance to ongoing antitumor therapy. The review reflects the immunological characteristics of CC, including in the context of molecu- lar biological subtypes. It describes the involvement of cells of the immune system (lymphocytes, macrophages) and their products (cytokines, chemokines) in the progression of colorectal cancer, including in the processes of neoangiogenesis, as well as the relationship of the T- and B-cell composition of the tumor microenvironment on the course of the disease. The review also shows the immunogenomic stratification of CC, which can be used to predict the response to immunotherapy for colorectal cancer.

For citation: Tishina A. V., Vladimirova L. Yu., Sagakyants A. B., Dzhenkova E. A., Novikova I. A., Zlatnik E. Yu. Immunologic aspects of colorectal cancer progression. South Russian Journal of Cancer. 2024; 5(1): 52-59. https://doi.org/10.37748/2686-9039-2024-5-1-6, https://elibrary.ru/wjwrxu

For correspondence: Anna V. Tishina - MD, oncologist of the Department of Oncohematology, National Medical Research Centre for Oncology, Rostov-on-Don, Russian Federation

Address: 63 14 line str., Rostov-on-Don 344037, Russian Federation ORCID: https://orcid.org/0000-0002-7990-8710

SPIN: 7686-3707, AuthorID: 965165

ResearcherID: H-2460-2018 Funding: this work was not funded

Conflict of interest: the authors declare that there are no obvious and potential conflicts of interest associated with the publication of this article

The article was submitted 06.12.2023; approved after reviewing 02.02.2024; accepted for publication 27.02.2024

????-?????????? ?????????????? ??????. 2024. ?. 5, ? 1. ?. 52-59 https://doi.org/10.37748/2686-9039-2024-5-1-6 https://elibrary.ru/wjwrxu

3.1.6. ?????????, ??????? ???????

?????

???????????????? ??????? ???????????????? ??????????????? ????

?. ?. ??????12, ?. ?. ???????????, ?. ?. ????????, ?. ?. ????????, ?. ?. ????????, ?. ?. ???????

???? "???????????? ??????????? ????????????????? ????? ?????????" ???????????? ??????????????? ?????????? ?????????, ?. ??????-??-????, ?????????? ?????????

12 tishina@sc-te.ru

?????????????? ??? ? ?????????? ?????????????? ? ?????????? ????? ????? ????? ??-???????? ???????? ?? ????- ?????? ????????. ??????? ?????????? ???????????? ???????? ?? ????????? ????? ???????????, ???????????? ??? ?? ?????? ???????????, ??? ? ?? ????????? ????????????? ????????????? ???????. ?????????????? ????????? ??????????? ?? ????? ???????????????, ???????????? ? ???????????????? ???????? ? ???????????? ????? ???????????? ???????????. ???????????? ???????????-???????????? ????????????? ? ??????????, ????????? ?????????????? ????????????? ???????, ????????????. ????? ??????????????? ??????? ????????? ?? ??????????- ?????? ????????? ????????? ? ??????? ?????? ???????? ???????, ?? ???????? ?????? ???? ?????????? ????????? ???????? ? ??????????????? ??????????, ? ????? ?????? ????????????????? ??????? ??????? ??????????????? ????. ???????????, ??? ????????? ???????? ?????? ? ?????????????? ??????? ???????? ????????????? ???????? ???????????????? ???????????. ???????? ? ???????? ? ?????????????? ??????? ??????????? ???????? ??????????, ? ?? ?????? ? ????????? ????? ???????? ??????? ?????????????? ??????? ? ?????????? ?????. ????????? ? ?????? ???????? ????????, ??????????? ? ????????? ???????????????? ? ?????????? ????????????????, ???????? ?????? ??????? ??????????? ????????. ????? ?????? ?????? ?????? ??????????? ????????????? ? ???????? ??????- ??????????? ??????????????, ? ???????????????? ??????????????? ????. ??????? ???????????? ?????????????? ??????? ?? ???????? ??????????, ? ????? ?? ?????????????? ? ?????????? ????????????????? ???????. ? ?????? ???????? ???????????????? ?????????????? ?????????????? ?????????, ? ??? ????? ? ????????? ???????????- ????????????? ????????. ??????????? ??????? ?????? ???????? ??????? (??????????, ??????????) ? ?? ????????? (?????????, ?????????) ? ???????????????? ??????????????? ????, ? ??? ????? ? ????????? ??????????????, ? ????? ??????????? ?- ? ?-?????????? ??????? ?????????????? ??????? ?? ??????? ???????????. ????? ? ?????? ?????????? ?????????????? ????????????? ?????????????? ?????????, ??????? ????? ???? ????????? ??? ??????????????? ?????? ?? ????????????? ??????????????? ????.

??? ???????????: ?????? ?. ?., ??????????? ?. ?., ???????? ?. ?., ???????? ?. ?., ???????? ?. ?., ??????? ?. ?. ???????????????? ??????? ???????????????? ??????????????? ????. ????-?????????? ?????????????? ??????. 2024; 5(1): 52-59. https://doi.org/10.37748/2686-9039-2024-5-1-6, https://elibrary.ru/wjwrxu

??? ???????????????: ?????? ???? ?????????? - ????-??????? ????????? ???????????????, ???? "???????????? ??????????? ????????????????? ????? ?????????" ???????????? ??????????????? ?????????? ?????????, ??????-??-????, ?????????? ????????? ?????: 344037, ?????????? ?????????, ?. ??????-??-????, ??. 14-? ?????, ?. 63

ORCID: https://orcid.org/0000-0002-7990-8710 SPIN: 7686-3707, AuthorID: 965165

ResearcherID: H-2460-2018

??????????????: ?????????????? ?????? ?????? ?? ???????????

???????? ?????????: ??? ?????? ???????? ?? ?????????? ????? ? ????????????? ?????????? ?????????, ????????? ? ??????????? ????????? ??????

?????? ????????? ? ???????? 06.12.2023; ???????? ????? ?????????????? 02.02.2024; ??????? ? ?????????? 27.02.2024

(c) ?????? ?. ?., ??????????? ?. ?., ???????? ?. ?., ???????? ?. ?., ???????? ?. ?., ??????? ?. ?., 2024

Colorectal cancer (CC) occupies a leading position in the structures of morbidity and mortality [1-6]. Despite the successes achieved in recent years in the diagnosis and therapy of cancer (targeted ther- apy, immunotherapy), the life expectancy of patients with this disease does not increase significantly. The reason for this may be the progression of the disease, as well as the development of resistance to therapy [7-9]. Molecular mechanisms of progression play a key role in metastasis [10].

To date, two classifications of colorectal cancer have been proposed reflecting the molecular genetic characteristics of the tumor [11-13]. In 2012, Can- cer Genome presented a molecular analysis of col- orectal carcinoma using genome-wide sequencing technology [11]. During the study, CC was divided into 2 groups, the first included tumors with a high mutational load or having microsatellite instability (MSI), the second group consisted of tumors with a low mutational load or having microsatellite sta- bility (MSS).

However, the criteria used in this classification turned out to be insufficient. During the data anal- ysis, new biomarkers of colorectal carcinoma were identified, which formed the basis of the new clas- sification. In 2016, Guinney et al., considering new data from the Consensus Molecular Subtype (CMS) consortium, the CC was divided into 4 subtypes (CMS1-CMS4) (Table. 1) [12]. The first subtype of CMS1 was characterized by the presence of MSI, the phenotype of methylation of CpG islands (CIMP), the presence of a mutation in the BRAF gene and

was called MSI - immune. The second subtype of CMS2 is canonical, characterized by the presence of a high level of somatic copies (SCNA), activation of MYC and the WNT signaling pathway. CMS3 or the third subtype is metabolic, it can include tumors with a mixed MSI status, low levels of SCNA and CIMP, and the presence of a mutation in the KRAS gene. The fourth CMS4 is mesenchymal, with the presence of high levels of SCNA, stromal infiltration, activation of TGF� and angiogenesis. At the same time, the authors not only classify colorectal carcinoma into certain subtypes, considering their molecular and genetic characteristics, but also give a prognosis regarding patient survival [12].

For example, patients with CMS1 are less like- ly to survive a relapse of the disease than patients with other subtypes, and patients with CMS4 have the worst prognosis for overall relapse-free survival compared to other subtypes.

However, this classification is not enough, since the cause of the progression CC is also associat- ed with the molecular heterogeneity of the tumor, which is part of the evolutionary and temporal pro- cess [14, 15]. Heterogeneity is also regarded as the cause of resistance to ongoing antitumor therapy (Fig. 1).

Tumor heterogeneity is often caused by a change in the RAS signaling pathway, which, in turn, is a com- ponent of the RAS-MEK-ERK cascade. Combinations of drugs, primarily anti-EGFR, are used to overcome resistance to EGFR inhibitors [16]. But even this ap- proach provides only a slight improvement in the

Table 1. Molecular subtypes of colorectal cancer [12] CMS1 Immune CMS2 (canonic) CMS3 (metabolic) CMS4 (mesenchymal) 14 % 37 % 13 % 23 % Increased expression of MSI Epithelial differentiation; Mixed status by MSI; Epithelial-mesenchymal genes;

High level of epithelial differentiation;

High mutational activity High somatic copyability Low level of epithelial differentiation;

Low somatic copyability transition;

High somatic copyability BRAF mutations KRAS mutations Immune infiltration Activation of the WNT and MYC signaling pathway Metabolic dysregulation Activation of TGF-b; Stromal infiltration; Angiogenesis Note: MSI - microsatellite instability; TGF - a transformative growth factor

survival rate of patients with metastatic CC. In order to find alternative ways to overcome resistance to ongoing therapy, as well as markers of drug efficacy, tumor genotyping based on blood samples is carried out, the effect of the immune system on tumor tissue is studied, including the search for new biomarkers. In the classification proposed by Guinney et al. [12] the immunological characteristics of CC are partially affected, in particular, the CMS1 subtype is charac- terized by the presence of infiltration of tumor stro- ma by immune cells. In addition, this subtype carries the ability to have a high level of mutational activity with the formation of neoantigens (resulting from somatic mutation of a tumor cell) that stimulate an antitumor immune response. This explains the high immunogenicity of the tumor and its infiltration by immune cells, especially activated lymphocytes - CD8+ T cells, CD4+ memory T cells, Th1, activated dendritic cells, NK cells and M1 macrophages. It is also known that CMS1 subtype tumors are able to express genes with subsequent release into the inter- cellular space of CXCL9 and CXCL10 involved in T cell chemotaxis, as well as IL-15, IFNy, CXCL13, etc. [17]. In addition, it has been shown that the expression of molecules of immune control points (PD-1, PD-L1, CTLA-4) of tumors of this subtype allows them to evade immune surveillance [18], although it suggests the effectiveness of immunotherapy with inhibitors of these control points in the treatment of such tumors. The CMS2 subtype is characterized by low levels

of lymphocytes, monocytes and myeloid cells, and, consequently, a weak antitumor response. In addi- tion, tumors of the "canonical" subtype practically do not express PD-1, PD-L1 [12].

Tumors belonging to the CMS3 subtype, as well as tumors with the CMS2 subtype, are characterized by an immunologically depleted cellular composition. However, unlike the previous subgroup, tumor cells carry PD-L1 on their surface, and there are Th17, "naive" B and T cells in their microenvironment [12, 19, 20]. Such a microenvironment, apparently, cannot provide an effective antitumor response, since Th17 has pro-oncogenic properties, and "naive" lympho- cytes do not have functional activity.

The fourth subgroup of colorectal carcinomas is characterized by a high level of infiltrating lym- phocytes and macrophages, with the M2 phenotype, while the number of M1 is reduced. A high content of regulatory T cells (T-reg) is also found, and the concentration of CD8+, CD4+ T cells is reduced. The presence of TGF-�, CXCL12 and VEGF contributes to the maintenance of the inflammatory environment and, as a result, causes the development and pro- gression of the tumor [12, 13, 19, 20].

A number of authors believe that, knowing the immunological, molecular and genetic component of various subtypes of colorectal cancer, it is possi- ble to predict the response to antitumor treatment [13, 19, 20].

Currently, immunological markers are being ac-

Fig. 1. Principles of evolutionary and temporal heterogeneity of cancer [14]

tively investigated as prognostic indicators of pro- gression [19-21], in particular, not only the type of immune cells infiltrating the tumor, which are part of the tumor microenvironment, but also the density of infiltration by these cells. At the same time, the approach to the study can be complex or multiplex and single-factor - the study of specific biomark- ers. The relevance of the study of immunological markers is due to the involvement of immune cells in the progression of cancer [22, 23]. Cytokines and chemokines both form an inflammatory environment and activate antitumor immunity. For example, IL-12, IL-15, IL-18, IFN-? stimulate the response to tumor antigens, and promote tumor progression - IL-6, IL-17A, IL-22, IL-23; affect neoangiogenesis, growth and survival of tumor cells - TNF-a, EGFR ligands, TGF-�, IL-6 [24]. Tumor-associated macrophages (TAM) play a key role in the development of both an inflammatory response and in the processes of progression and are also a source of a wide range of cytokines.

Macrophages are the most common immune cells

in the microenvironment of colorectal carcinoma. Macrophages are able not only to influence the pro- cesses of inflammation in the microenvironment, but also participate in carcinogenesis and tumor progres- sion. In addition, they can modulate the response to standard treatment methods (chemotherapy, radiation therapy, therapy with drugs suppressing

neoangiogenesis), leading to the development of re- sistance and subsequent tumor progression [25-28]. For example, the expression of IL-6 and TNF-a mac- rophages promotes the transmission of signals by tumor cells and the development of resistance to antitumor therapy. The invasion of neoplastic cells is facilitated by the targeted release of cytokines/ chemokines, such as EGF, CCCL18, IL-4. Macro- phages participate in the processes of neoangio- genesis by stimulating the expression of VEGF-A by endothelial cells, which in turn leads to the for- mation of an abnormal vascular network, which is characterized by excessive branching, a large num- ber of capillaries, lack of vascular tightness, thereby changing hemodynamics in tumor tissue, making it difficult to deliver drugs. Macrophages are also able to influence cytotoxic lymphocytes by modulating the immune response. The inhibition of the cytotoxic T lymphocyte response can occur through the expres- sion of B7 family ligands or by the release of IL-10 through CCL22 with suppression of IL-12 production by dendritic cells.

The immunosuppressive role is played by regula-

tory T cells (T-reg) due to the production of anti-in- flammatory cytokines IL-10 and TGF-� [29]. B-cell infiltration in CC is often observed due to the large representation of these cells in tertiary lymphoid structures that originate from peripheral lymphoid tissue under prolonged exposure to inflammatory

Fig. 2. Cluster typing of the immune response (CIRC)

Table 2. Genes of clusters coordinating the immune response [33] Group Genes Group ? HLA-DQA1 HLA-DQA2 HLA-DRB5 HLA-DMA PDCD1LG2 ICAM1 CD274 Group ? STAT1 IRF1 IFNG CTLA4 TBX21 CCL5 LAG-3 Group ? CD247 ICOS IL18RAP GNLY CXCL10 HLA-DPB1 HLA-DPA1 Group D HLA-DMB HLA-DRA HLA-DMA CD80 HLA-DOA CD4 HAVCR2

signals mediated by chemokines and cytokines [30]. B cells in the tumor microenvironment along with the T-cell component (cytotoxic CD3+CD4+ and CD3+CD8+ T cells, other subpopulations of T cells) are associated with a favorable prognosis. However, the presence of macrophages in the CC microenvi- ronment stimulates the development of inflammation and, as a result, affects tumor progression [31].

The phenomena occurring in immunocompetent cells of the CC microenvironment may also differ at the molecular genetic level. Thus, Laghi L., et al., in 2020 published a paper aimed at identifying the relationship between the genetic and immune com- ponents of colorectal cancer [32]. It is known that tumors with MSI have a large number of tumor in- filtrating lymphocytes (TILs), however, tumors with MSS may also have high levels of TILs. A favorable prognosis in CC is associated with a high level of TILs, which in turn can be a biomarker for identifying a cohort of patients with a low probability of disease recurrence and influence the choice of therapy.

The search for biomarkers capable of predicting the effectiveness of therapy in CC continues. Lal N, et al., published a paper on the immunogenomic strat- ification of colorectal carcinoma used to describe the response to CC immunotherapy [33]. The basis for stratification was cluster typing of the immune response (CIRC) (Fig. 2) [33], which divides patients with CC into four groups depending on the level of ex- pression of a set of genes that do not completely co- incide with the molecular genetic subtypes (Table 2).

Stratification links the genetics and immunobiol- ogy of CC. At the same time, the expression of im- mune control points and cytokine/chemokine genes were found to correspond to the expression of some variants of the main histocompatibility complex HLA (Table 2).

Group A is characterized by MSI-H and POLE gene mutations, high mutational load and high immune infiltration, which can be useful when using immune checkpoint inhibitors (ICIs). Whereas in group D and B, mutations in the RAS family genes were present, and these patients were resistant to ICI therapy. Nev- ertheless, the question remains which of the CC clas- sifications to focus on when predicting the response to, particularly, immunotherapy treatment [34]. The development of new approaches to the stratifica- tion of patients with CC continues, as well as the search for new directions to eliminate resistance in the population of patients resistant to existing treatment methods.

CONCLUSION

The tumor microenvironment by immune cells plays one of the key roles in the progression of colorectal cancer and the mechanisms of develop- ment of resistance to therapy, which may be sig- nificant for a personalized approach to antitumor treatment and the search for predictive markers of the effectiveness of therapy, including immu- nological ones.

References

1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021 Jan;71(1):7-33. https://doi.org/10.3322/caac.21654

2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Es- timates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021 May;71(3):209-249. https://doi.org/10.3322/caac.21660

3. Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gas- troenterol. 2019;14(2):89-103. https://doi.org/10.5114/pg.2018.81072

4. Osombaev MSh, Dzhekshenov MD, Satybaldiev OA, Abdrasulov KD, Makimbetov EK, Kuzikeev MA. Epidemiology of colorectal cancer. Scientific review. Medical Sciences. 2021;1(7):37-42. (In Russ.). https://doi.org/10.17513/srms.1169, EDN: LYEMDE

5. Vodolazhsky DI, Antonets AV, Dvadnenko KV, Vladimirova LYu, Gevorkyan YuA, Kasatkin VF, et al. The relationship of KRAS gene mutations with the clinical and pathological features of colorectal cancer in patients in the South of Russia. Interna- tional Journal of Experimental Education. 2014;1(1):65-68. (In Russ.).

6. The state of cancer care for the Russian population in 2021. Edited by A. D. Kaprin, V. V. Starinsky, A. O. Shakhzadova. Mos- cow: P. A. Herzen MNIOI - Branch of the National Medical Research Radiological Center, 2022, 239 p. (In Russ.)

7. Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020 Mar 20;5(1):22. https://doi.org/10.1038/s41392-020-0116-z

8. Giordano G, Remo A, Porras A, Pancione M. Immune Resistance and EGFR Antagonists in Colorectal Cancer. Cancers (Ba- sel). 2019 Jul 31;11(8):1089. https://doi.org/10.3390/cancers11081089

9. Woolston A, Khan K, Spain G, Barber LJ, Griffiths B, Gonzalez-Exposito R, et al. Genomic and Transcriptomic Determinants of Therapy Resistance and Immune Landscape Evolution during Anti-EGFR Treatment in Colorectal Cancer. Cancer Cell. 2019 Jul 8;36(1):35-50.e9. https://doi.org/10.1016/j.ccell.2019.05.013

10. Malki A, ElRuz RA, Gupta I, Allouch A, Vranic S, Al Moustafa AE. Molecular Mechanisms of Colon Cancer Progression and Metastasis: Recent Insights and Advancements. Int J Mol Sci. 2020 Dec 24;22(1):130. https://doi.org/10.3390/ijms22010130

11. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012 Jul 18;487(7407):330-337. https://doi.org/10.1038/nature11252

12. Guinney J, Dienstmann R, Wang X, de Reyni�s A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of col- orectal cancer. Nat Med. 2015 Nov;21(11):1350-1356. https://doi.org/10.1038/nm.3967

13. Wang Y, Han G, Wang K, Liu G, Wang R, Xiao H, et al. Tumor-derived GM-CSF promotes inflammatory colon carcinogenesis via stimulating epithelial release of VEGF. Cancer Res. 2014 Feb 1;74(3):716-726.

https://doi.org/10.1158/0008-5472.CAN-13-1459

14. Gaiani F, Marchesi F, Negri F, Greco L, Malesci A, de'Angelis GL, et al. Heterogeneity of Colorectal Cancer Progression: Mo- lecular Gas and Brakes. Int J Mol Sci. 2021 May 15;22(10):5246. https://doi.org/10.3390/ijms22105246

15. Hunter KW, Amin R, Deasy S, Ha NH, Wakefield L. Genetic insights into the morass of metastatic heterogeneity. Nat Rev Cancer. 2018 Apr;18(4):211-223. https://doi.org/10.1038/nrc.2017.126

16. Reutova EV, Laktionov KK, Ardzinba MS, Nelyubina LA, Arzumanyan AL. Acquired resistance to EGFR tyrosine kinase inhib- itors: ways to overcome. Medical Council. 2017;(14):24-28. (In Russ.). https://doi.org/10.21518/2079-701X-2017-14-24-28

17. Becht E, de Reyni�s A, Giraldo NA, Pilati C, Buttard B, Lacroix L, et al. Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy. Clin Cancer Res. 2016 Aug 15;22(16):4057-4066. https://doi.org/10.1158/1078-0432.CCR-15-2879

18. Karpinski P, Rossowska J, Sasiadek MM. Immunological landscape of consensus clusters in colorectal cancer. Oncotar- get. 2017 Dec 1;8(62):105299-105311. https://doi.org/10.18632/oncotarget.22169

19. Piawah S, Venook AP. Targeted therapy for colorectal cancer metastases: A review of current methods of molecularly targeted therapy and the use of tumor biomarkers in the treatment of metastatic colorectal cancer. Cancer. 2019 Dec 1;125(23):4139-4147. https://doi.org/10.1002/cncr.32163

20. Okita A, Takahashi S, Ouchi K, Inoue M, Watanabe M, Endo M, et al. Consensus molecular subtypes classification of col- orectal cancer as a predictive factor for chemotherapeutic efficacy against metastatic colorectal cancer. Oncotarget. 2018 Apr 10;9(27):18698-18711. https://doi.org/10.18632/oncotarget.24617

21. Zafari N, Khosravi F, Rezaee Z, Esfandyari S, Bahiraei M, Bahramy A, et al. The role of the tumor microenvironment in col- orectal cancer and the potential therapeutic approaches. J Clin Lab Anal. 2022 Aug;36(8):e24585. https://doi.org/10.1002/jcla.24585

22. Nikipelova EA, Kit OI, Shaposhnikov AV, Zlatnik EYu, Novikova IA, Vladimirova LYu, et al. Immunological criteria for the devel- opment of distant metastases of colon cancer. News of higher educational institutions. The North Caucasus region. Series: Natural Sciences. 2017;(3-2(195-2)):96-101. (In Russ.). https://doi.org/10.23683/0321-3005-2017-3-2-96-101, EDN: ZQTDAB

23. Kit OI, Dzhenkova EA, Mirzoyan EA, Sagakyants AB, Bondarenko ES, Zlatnik EYu, et al. Features of local cellular immunity in colon cancer, depending on the localization of the tumor process. Modern Problems of Science and Education. 2022;3:86. (In Russ.). https://doi.org/10.17513/spno.31695

24. Mager LF, Wasmer MH, Rau TT, Krebs P. Cytokine-Induced Modulation of Colorectal Cancer. Front Oncol. 2016 Apr 19;6:96. https://doi.org/10.3389/fonc.2016.00096

25. Ruffell B, Coussens LM. Macrophages and therapeutic resistance in cancer. Cancer Cell. 2015 Apr 13;27(4):462-472. https://doi.org/10.1016/j.ccell.2015.02.015

26. Coffelt SB, de Visser KE. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol. 2015 Apr;36(4):198-216. https://doi.org/10.1016/j.it.2015.02.006

27. Engblom C, Pfirschke C, Pittet MJ. The role of myeloid cells in cancer therapies. Nat Rev Cancer. 2016 Jul;16(7):447-462. https://doi.org/10.1038/nrc.2016.54

28. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014 Jul 17;41(1):49-61. https://doi.org/10.1016/j.immuni.2014.06.010

29. Whiteside TL. What are regulatory T cells (Treg) regulating in cancer and why? Semin Cancer Biol. 2012 Aug;22(4):327-334. https://doi.org/10.1016/j.semcancer.2012.03.004

30. Saut�s-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019 Jun;19(6):307-325. https://doi.org/10.1038/s41568-019-0144-6

31. Liu Q, Yang C, Wang S, Shi D, Wei C, Song J, et al. Wnt5a-induced M2 polarization of tumor-associated macrophages via IL-10 promotes colorectal cancer progression. Cell Commun Signal. 2020 Mar 30;18(1):51. https://doi.org/10.1186/s12964-020-00557-2

32. Laghi L, Negri F, Gaiani F, Cavalleri T, Grizzi F, De' Angelis GL, et al. Prognostic and Predictive Cross-Roads of Microsatellite Instability and Immune Response to Colon Cancer. Int J Mol Sci. 2020 Dec 18;21(24):9680. https://doi.org/10.3390/ijms21249680

33. Lal N, Beggs AD, Willcox BE, Middleton GW. An immunogenomic stratification of colorectal cancer: Implications for devel- opment of targeted immunotherapy. Oncoimmunology. 2015 Mar;4(3):e976052. https://doi.org/10.4161/2162402X.2014.976052

34. Zheng X X, Ma Y, Bai Y, Huang T, Lv X, Deng J, et al. Identification and validation of immunotherapy for four novel clusters of colorectal cancer based on the tumor microenvironment. Front Immunol. 2022;13:984480. https://doi.org/10.3389/fimmu.2022.984480

Information about authors:

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

Anna V. Tishina 12 - MD, oncologist of the Department of Oncohematology, National Medical Research Centre for Oncology, Rostov-on-Don, Russian

Federation

ORCID: https://orcid.org/0000-0002-7990-8710, SPIN: 7686-3707, AuthorID: 965165, ResearcherID: H-2460-2018

Liubov Yu. Vladimirova - Dr. Sci. (Med.), professor, head of the Department of Antitumor Drug Therapy, head of the Department of Drug Treatment of Tumors, National Medical Research Centre for Oncology, Rostov-on-Don, Russian Federation

ORCID: https://orcid.org/0000-0002-4822-5044, SPIN: 4857-6202, AuthorID: 289090, ResearcherID: U-8132-2019, Scopus Author ID: 7004401163

Aleksandr B. Sagakyants - Dr. Sci. (Biol.), associate professor, head of the laboratory of Tumor Immunophenotyping, National Medical Research Centre for Oncology, Rostov-on-Don, Russian Federation

ORCID: https://orcid.org/0000-0003-0874-5261, SPIN: 7272-1408, AuthorID: 426904, ResearcherID: M-8378-2019, Scopus Author ID: 24329773900

Elena A. Dzhenkova - Dr. Sci. (Biol.), professor, academic secretary, National Medical Research Centre, Rostov-on-Don, Russian Federation ORCID: https://orcid.org/0000-0002-3561-098X, SPIN: 6206-6222, AuthorID: 697354, ResearcherID: K-9622-2014, Scopus Author ID: 6507889745

Inna A. Novikova - Dr. Sci. (Med.), deputy director for science, National Medical Research Centre for Oncology, Rostov-on-Don, Russian Federation ORCID: https://orcid.org/0000-0002-6496-9641, SPIN: 4810-2424, AuthorID: 726229, ResearcherID: E-7710-2018, Scopus Author ID: 7005153343

Elena Yu. Zlatnik - Dr. Sci. (Med.), professor, chief researcher of the Laboratory of Tumor Immunophenotyping, National Medical Research Centre of Oncology, Rostov-on-Don, Russian Federation

ORCID: https://orcid.org/0000-0002-1410-122X, SPIN: 4137-7410, AuthorID: 327457, ResearcherID: AAI-1311-2020, Scopus Author ID: 6603160432

Contribution of the authors:

Dzhenkova E. A., Vladimirova L. Yu. - scientific guidance, final conclusions; Zlatnik E. Yu., Tishina A. V. - writing the primary text;

Sagakyants A. B., Novikova I. A. - revision and correction of the text, final conclusions.

i Надоели баннеры? Вы всегда можете отключить рекламу.