Научная статья на тему 'NIVOLUMAB MONOTHERAPY IN METASTATIC COLORECTAL CANCER: CURRENT APPROACHES TO RESPONSE EVALUATION'

NIVOLUMAB MONOTHERAPY IN METASTATIC COLORECTAL CANCER: CURRENT APPROACHES TO RESPONSE EVALUATION Текст научной статьи по специальности «Клиническая медицина»

CC BY
22
6
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
METASTATIC COLORECTAL CANCER / NIVOLUMAB / ATYPICAL RESPONSE PATTERNS

Аннотация научной статьи по клинической медицине, автор научной работы — Babyshkina N.N., Dronova T.A., Gervas P.A., Popova N.O., Dobrodeev A.Yu.

Aim of the study: a systematic analysis of the modern literature data on the nivolumab monotherapy efficacy in patients with metastatic colorectal cancer (mCRC). Material and Methods. The review summarizes the results of clinical studies of the nivolumab efficacy in patients with mCRC between 2012 and 2022. The current approaches to assessing the tumor response in patients treated with immune checkpoint inhibitors are considered, including response patterns and criteria. Results. Data analysis showed that the use of nivolumab in mCRC patients had significant clinical benefits. Nivolumab monotherapy was shown to improve survival in patients with high microsatellite instability (MSi) or deficiencies in mismatch repair (dMMR) that progressed during standard chemotherapy. Numerous clinical studies indicate the atypical response to nivolumab. Traditional response criteria, such as RECiST do not always adequately assess the therapeutic efficacy of nivolumab in patients with mCRC. conclusion. To improve the efficacy of mCRC treatment, standardized approaches based on the proposed specific criteria for response to immunotherapy, including immune related RECiST, immune RECiST, and immune-modified RECiST must be developed.

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

Текст научной работы на тему «NIVOLUMAB MONOTHERAPY IN METASTATIC COLORECTAL CANCER: CURRENT APPROACHES TO RESPONSE EVALUATION»

DOI: 10.21294/1814-4861-2022-21-5-135-141 УДК: 616.34-006.6-003.2-08:615.37

Для цитирования: Бабышкина Н.Н., Дронова Т.А., Гервас П.А., Попова Н.О., Добродеев А.Ю., Костро-мицкий Д.Н., Афанасьев С.Г., Гольдберг В.Е., Чердынцева Н.В. Монотерапия ниволумабом у больных метастатическим колоректальным раком: современные подходы к оценке ответа опухоли. Сибирский онкологический журнал. 2022; 21(5): 135-141. - doi: 10.21294/1814-4861-2022-21-5-135-141

For citation: Babyshkina N.N., Dronova T.A., Gervas P.A., Popova N.O., DobrodeevA.Yu., KostromitskiyD.N., Afa-nasyev S.G., Goldberg V.E., Cherdyntseva N.V. Nivolumab monotherapy in metastatic colorectal cancer: current approaches to response evaluation. Siberian Journal of Oncology. 2022; 21(5): 135-141. - doi: 10.21294/1814-4861-2022-21-5-135-141

nivolumab monotherapy in metastatic colorectal cancer: current approaches to response evaluation

N.N. Babyshkina12, T.A. Dronova1, p.A. Gervas1, N.o. popova1, A.Yu. dobrodeev1, D.N. Kostromitskiy1, s.G. Afanasyev1, V.E. Goldberg1, N.V. cherdyntseva1

Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia1

5, Kooperativny St., 634009, Tomsk, Russia. E-mail: nbabyshkina@mail.ru1 Siberian State Medical University of the Ministry of Health of Russia, Tomsk, Russia2 2, Moskovsky tract, 634050, Tomsk, Russia2

Abstract

Aim of the study: a systematic analysis of the modern literature data on the nivolumab monotherapy efficacy in patients with metastatic colorectal cancer (mCRC). Material and Methods. The review summarizes the results of clinical studies of the nivolumab efficacy in patients with mCRC between 2012 and 2022. The current approaches to assessing the tumor response in patients treated with immune checkpoint inhibitors are considered, including response patterns and criteria. Results. Data analysis showed that the use of nivolumab in mCRC patients had significant clinical benefits. Nivolumab monotherapy was shown to improve survival in patients with high microsatellite instability (MSI) or deficiencies in mismatch repair (dMMR) that progressed during standard chemotherapy. Numerous clinical studies indicate the atypical response to nivolumab. Traditional response criteria, such as RECIST do not always adequately assess the therapeutic efficacy of nivolumab in patients with mCRC. conclusion. To improve the efficacy of mCRC treatment, standardized approaches based on the proposed specific criteria for response to immunotherapy, including immune related RECIST, immune RECIST, and immune-modified RECIST must be developed.

Key words: metastatic colorectal cancer, nivolumab, REOisT, atypical response patterns.

монотерапия ниволумабом у больных метастатическим колоректальным раком: современные подходы к оценке ответа опухоли

Н.Н. Бабышкина12, Т.А. Дронова1, П.А. Гервас1, Н.О. Попова1,

А.Ю. Добродеев1, Д.Н. Костромицкий1, С.Г. Афанасьев1, В.Е. Гольдберг1,

Н.В. Чердынцева1

Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук, г Томск, Россия1 Россия, г. Томск, 634009, пер. Кооперативный, 5. E-mail: nbabyshkina@mail.ru1

ФГБОУ ВО «Сибирский государственный медицинский университет» Минздрава России, г. Томск, Россия2

Россия, г Томск, 634050, Московский тракт, 222 1=7 Бабышкина Наталия Николаевна, nbabyshkina@mail.ru

Цель исследования - систематический анализ данных современной литературы об эффективности применения ниволумаба в монорежиме у больных метастатическим колоректальным раком (мКРР). Материал и методы. В обзор включены данные клинических исследований за период с 2012 по 2022 г, оценивающих эффективность лечения ниволумабом у больных мКРР Рассмотрены текущие подходы к оценке опухолевого ответа, включая характер иммунного ответа и критерии ответа. Результаты. Анализ литературы показал, что применение ниволумаба при мКРР имеет значимые клинические преимущества. Продемонстрировано, что монотерапия ниволумабом улучшает показатели выживаемости у пациентов с высокой микросателлитной нестабильностью (М51) или нарушением механизма репарации неспаренных оснований ДНК (dММR), которые прогрессировали на фоне стандартной химиотерапии. Многочисленные клинические исследования указывают на развитие атипичных реакций ответа на ниволумаб. Традиционные критерии ответа, такие как RECIST, не всегда адекватно оценивают терапевтическую эффективность ниволумаба у пациентов с мКРР. Заключение. Для повышения эффективности лечения мКРР необходима разработка стандартизированных подходов, основанных на предложенных специфических критериях ответа на иммунотерапию, включая иммунологический RECIST, иммунный RECIST и иммуномодифицированный RECIST

Аннотация

Ключевые слова: метастатический колоректальный рак, ниволумаб, RECiST, атипичный характер ответа.

Introduction

Colorectal cancer (CRC) is the leading cause of cancer-related death, both worldwide and in the Russian Federation [1-2]. The metastatic CRC (mCRC) represents approximately 30-50 % of all initially detected disease cases [3, 4]. Current treatment strategies of CRC are based on a multimodal approach, including chemotherapy, targeted therapy and immunotherapy, which allow selection of the optimal individualized treatment.

According to the ESMO, NCCN recommendations, as well as the Ministry of Health of the Russian Federation guidance, screening for high microsatellite instability (MSI) and deficiencies in mismatch repair (dMMR) is recommended to all mCRC patients for prediction of clinical benefit from immunotherapy. Currently, three immune checkpoint inhibitors, such as ipilimumab, pembrolizumab, and nivolumab are approved for use in MSI-high and dMMR advanced CRC patients [5-7]. It was demonstrated that nivolumab improved objective response rate and progression-free survival with a manageable safety profile compared to standard therapy for melanoma, renal cell carcinoma, and non-small cell lung cancer [8-10]. Further studies have confirmed that nivolumab provides a durable response and disease control in pre-treated mCRC patients, including patients with dMMR/MSI-high tumor [11-13]. In this review, we summarize the clinical aspects of nivolumab administration, mainly focusing on tumor response patterns in mCRC patients and current approaches to its assessing.

Nivolumab action and safety

Nivolumab has been developed in research collaboration between Ono Pharmaceutical and Me-darex Company with the originally name MDX-1106/ ONO-4538, This is a human IgG4 monoclonal antibody that contains a hinge region mutation (S228P),

which reduces Fc exchange with IgG4 molecules to improve stability and reduce therapeutic variability

[14]. Nivolumab blocks the interaction between the programmed cell death 1 (PD-1) receptor and PD-L1/ PD-L2 ligands. Inhibition of PD-1 and its ligands promotes the reactivation of tumor-specific T-lymphocytes and following prolongation of their antitumor effect

[15]. Pharmacokinetic studies have suggested that nivolumab has linear pharmacokinetics with a doseproportional increase in the maximum concentration and area under the concentration-time curve. The time to peak plasma nivolumab concentration ranges between 1-4 hours [14]. In August 2017, the US Food and Drug Administration (FDA) approved nivolumab for the treatment of patients with dMMR/MSI-high mCRC that had progressed following chemotherapy. Nivolumab is administered at dose 240 mg as an intravenous infusion over 30 minutes every 2 weeks until disease progression or unacceptable toxicity.

In more than 20 % of patients receiving nivolumab as monotherapy, the most common observed adverse reactions are fatigue, rash, musculoskeletal pain, pruritus, diarrhea, nausea, asthenia, cough, dyspnea, constipation, decreased appetite, back pain, arthralgia, upper respiratory tract infection, and pyrexia. The main side effects of nivolumab monotherapy leading to discontinuation of treatment are diarrhea or colitis grade 4, pneumonitis grade 3 or 4, AST or ALT >5 times ULN or total bilirubin >3 times ULN, hypophysitis grade 4, adrenal insufficiency 3 or 4 grade 4 hyperglycemia, serum creatinine >6 times ULN, grade 4 rash or confirmed Stevens-Johnson syndrome or toxic epidermal necrolysis, immune-mediated encephalitis, recurrent grade 3 adverse reactions, life-threatening or adverse reactions 4 grade, need for >10 mg/day prednisone or equivalent for >12 weeks, and persistent grade 2 or 3 adverse reactions lasting > 12 weeks. No specific for mCRC side effects have been identified [16].

Response patterns and criteria

RECIST assessment

Radiologic response evaluation using the Response Evaluation Criteria in Solid Tumors (RECIST V.1.1) with assessments of complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD) remains the standard option in routine clinical practice. Complete response to nivolumab persisting for 21 months in a 67-year-old male with CRC with metastases in intra-abdominal lymph nodes was reported for the first time in 2010 [14]. In a subsequent multicenter phase 1 trial, nivolumab did not show clinical activity in mCRC patients [17]. However, one prolonged (for >6 months) partial response to nivolumab in a CRC patient was observed at a single-center, open label Japanese study [13]. In addition, two complete responses and seven partial responses in patients with treatment-refractory meta-static anal cancer were reported in a multicentre, phase 2 trials from ten USA academic centers. Among nine responders, seven patients had a durable response with a median duration of response of 5.8 months [12]. With the evidence for the heterogeneous nature of cancer; it has become clear that PD-1/PDL-1 targeting could be effective in selected cohort of patients. This was confirmed by CheckMate 142 multicentre phase 2 study that nivolumab had encouraging efficacy in patients with dMMR/MSI-high mCRC [11]. According to his assessment, the objective response rate was achieved in 23 patients (31.1 %) and disease control for >12 weeks was observed in 51 patients (68.9 %). In addition, few reports have been demonstrated cases with a complete clinical response to nivolumab, including a patient with metastatic clear cell renal cell carcinoma [18] and a patient with gastric cancer [19].

Atypical response patterns

Recently, new atypical response patterns have been described in patients treated with immune checkpoint inhibitors that are not seen with chemotherapy and targeted therapy. These include the durable response, pseudoprogression, hyperprogression, and dissociated response [20-21].

Durable response is defined as a treatment response for an extended time even after stopping medication. To date, only one durable response to nivolumab (longer than 36 months) has been reported in a 71-year-old male with MSI-high mCRC [22]. The same response to nivolumab (range 8.07+—48.0+) was described in patients with metastatic papillary renal cell carcinoma [23], metastatic HPV-positive head and neck squamous cell carcinoma [24] and metastatic melanoma [25]. However, no durable response criteria have been developed to date.

The phenomenon of pseudoprogression was described for first time in advanced melanoma patients treated with pembrolizumab [26]. It has been identified as tumor shrinkage after an initial increase in tumor burden or the appearance of new lesions [27]. Pseudo-

progression represents a rare response type. One study reported no pseudoprogression in patients subgroup with MSI-high mCRC treated with anti-PD-1 antibody [28]. Chae et al. demonstrated a pseudoprogression during combinational treatment of PD-L1 inhibitor and 0X40 agonist in MSI-high metastatic colorectal adenocarcinoma patient who experienced initial increase in size and number of tumor lesions with subsequent tumor regression. An extraordinary scale of pseudoprogression with 163 % tumor growth from baseline was reported [29]. It has been recently shown that pseudoprogression occurs in 10 % of patients with dMMR/MSI-high mCRC treated with immune checkpoint inhibitors, including nivolumab monotherapy. Pseudoprogression is more frequently observed in patients treated with anti-PD1 monotherapy than in those receiving combination of anti-PD1 and anti-CTLA-4. In addition, 8 of the 12 patients with an initial pseudoprogression achieved an objective response (five partial responses and three complete responses) and had favorable outcomes [30].

Hyperprogression response is characterized by rapid tumor progression, which often leads to unfavorable outcome. The concept of hyperprogression remains controversial since there is no substantiated evidence of its occurrence under therapy with immune checkpoint inhibitors. In addition, assessment criteria of hyperprogression based on tumor growth kinetics are highly heterogeneous [20]. However, some case reports on hyperprogression under pembrolizumab have been documented in the literature, particularly in patients with metastatic MSI-high/dMMR CRC [31-33], and advanced colon cancer with Lynch syndrome [34]. With regard to nivolumab, one case of a dMMR gastrointestinal cancer patient who experienced hyperprogressive disease following its administration was presented. In this case report, 1.5 month after starting nivolumab (one dose of nivolumab), the patient experienced rapid metastatic progression in the lungs and lymph nodes and patient died of respiratory failure [35].

A dissociated response is observed when some lesions shrink and existing lesions enlarge or new lesions appear. This term is not well-established; terms such as mixed or heterogeneous response are used to define this response. Various types of dissociated response under nivolumab were described in patients with advanced cancers [21, 22, 36-39]. There are no reported cases of this response type in patients with metastatic MSI-high/dMMR CRC. However, we have our experience of observing a dissociated response in a patient with MSI-high metastatic colorectal cancer after 8 months of treatment with nivolumab with reduced pulmonary nodules and concomitant para-aortic lymph node involvement.

Immune-related response criteria

To better characterize the atypical response patterns, different immune-related response criteria based

on new approaches to PR and PD evaluation were developed. Immune-related response criteria (irRC) have become the first tool to use a bi-dimensional measurement of tumor lesions, when new lesions are not considered as PD and incorporated into the sum of measurements of the total tumor burden. This criterion requires the confirmation of progression at least 4 weeks after initial assessment [40]. Subsequently, irRC became the background of REClST-based immune criteria (ir-RECIST). In fact, ir-RECIST uses the same scores as RECIST V.1.1. According to ir-RECIST, the definition of complete response is disappearance of all lesions, partial response defined as >30 % reduction tumor burden from baseline, absence of both CR and PD are stable disease, and progression is a 20 % increase in total measurable tumor mass from a nadir of at least 5 mm, progression of non-target lesions or the appearance of a new lesion, which must also be confirmed at least 4 weeks and up to 12 weeks after initial assessment [41]. The next step was the approval of a modified immune RECIST criterion (iRECIST), with new responses terminology for CR (iCR), SD (iSD), PR (iPR), and unconfirmed PD (iUPD) or confirmed PD (iCPD). In addition, new lesions assessed as per RECIST V.1.1 but recorded separately on the case report form (but not included in the sum of lesions for target lesions identified at baseline) [42]. Finally,

ЛИТЕРАТУРА/REFERENCES

1. SungH., Ferlay J., SiegelR.L., LaversanneM., Soerjomataram I., Jemal A., Bray F. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71(3): 209-49. doi: 10.3322/caac.21660.

2. Злокачественные новообразования в России в 2020 году (заболеваемость и смертность). Под ред. А.Д. Каприна, В.В. Старин-ского, А.О. Шахзадовой. М., 2021. 252 с. [Malignant tumors in Russia in 2020 (morbidity and mortality). Ed. by A.D. Kaprin, V.V. Starinsky, A.O. Shakhzadova. Moscow, 2021. 252 p. (in Russian)].

3. Aparicio J., Esposito F., Serrano S., Falco E., Escudero P., Ruiz-CasadoA., ManzanoH. Fernandez-Montes A. Metastatic colorectal cancer. First line therapy for unresectable disease. J Clin Med. 2020; 9(12): 3889. doi: 10.3390/jcm9123889.

4. Одинцова И.Н., Черемисина О.В., Писарева Л.Ф., Спивакова И.О., Вусик М.В. Эпидемиология колоректального рака в Томской области. Сибирский онкологический журнал. 2017; 16(4): 89-95. [Odin-tsova I.N., Cheremisina O.V., Pisareva L.F., Spivakova I.O., VusikM.V. Epidemiology of colorectal cancer in Tomsk region. Siberian Journal of Oncology. 2017; 16(4): 89-95. (in Russian)]. doi: 10.21294/1814-48612017-16-4-89-95.

5. BensonA.B., Venook A.P., Al-HawaryM.M., ArainM.A., Chen Y.J., Ciombor K.K., Cohen S., Cooper H.S., Deming D., Farkas L., Garrido-Laguna I., Grem J.L., Gunn A., Hecht J.R., Hoffe S., Hubbard J., Hunt S., Johung K.L., Kirilcuk N., Krishnamurthi S., Messersmith W.A., Meyerhardt J., Miller E.D., Mulcahy M.F., Nurkin S., OvermanM.J., Parikh A., Patel H., Pedersen K., Saltz L., Schneider C., Shibata D., Skibber J.M., Sofocleous C.T., Stoffel E.M., Stotsky-Himelfarb E., Willett C.G., Gregory K.M., Gurski L.A. Colon cancer, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2002; 19(3): 329-59. doi: 10.6004/jnccn.2021.0012.

6. Van Cutsem E., Cervantes A., Adam R., Sobrero A., Van Krieken J.H., Aderka D., Aranda Aguilar E., Bardelli A., Benson A., Bodoky G., Ciardiello F., D'Hoore A., Diaz-Rubio E., Douillard J.Y., Ducreux M., Falcone A., Grothey A., Gruenberger T., Haustermans K., Heinemann V., Hoff P., Kohne C.H., Labianca R., Laurent-Puig P., Ma B., Maughan T., Muro K., Normanno N., OsterlundP., Oyen W.J., Papamichael D., Pen-theroudakis G., Pfeiffer P., Price T.J., Punt C., Ricke J., Roth A., Salazar R., Scheithauer W., Schmoll H.J., Tabernero J., Taieb J., Tejpar S., Wasan H., Yoshino T., Zaanan A., Arnold D. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016; 27(8): 1386-1422. doi: 10.1093/annonc/mdw235.

the next modification of immune-modified RECIST (imRECIST) compared to RECIST V.1.1 included allowance for best overall response after progressive disease and changes in PD definitions per new lesions and non-target lesions. imRECIST progression-free survival did not count initial PD as an event if the subsequent scan showed disease control [43].

All new immune-related response criteria are actively used in clinical trials of immune checkpoint inhibitors. However, none of these has been adapted for routine clinical practice.

Conclusion and future perspectives

Nivolumab monotherapy in metastatic colorectal cancer was shown to provide clinical benefit and improves survival in patients with dMMR/MSI-high mCRC who progressed after receiving a standard chemotherapy. Numerous clinical studies suggest that nivolumab may lead to atypical response patterns. Traditional response criteria such as RECIST may not always adequately assess the therapeutic efficacy of nivolumab in patients with metastatic colorectal cancer. To improve the management of mCRC, it is important to develop standardized approaches based on the proposed specific criteria for response to im-munotherapy, including immune related RECIST, immune RECIST and immune-modified RECIST.

7. Злокачественные новообразования ободочной кишки и ректосигмоидного отдела. МКБ 10: C18, C19. 2020. [Cancer of the colon and rectosigmoid. ICD 10: C18, C19. 2020. (in Russian)].

8. TopalianS.L., SznolM., McDermott D.F., KlugerH.M., CarvajalR.D., Sharfman W.H., Brahmer J.R., LawrenceD.P., AtkinsM.B., Powderly J.D., Leming P.D., Lipson E.J., Puzanov I., Smith D.C., Taube J.M., Wiggin-ton J.M., Kollia G.D., Gupta A., Pardoll D.M., Sosman J.A., Hodi F.S. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014; 32(10): 1020-30. doi: 10.1200/JC0.2013.53.0105.

9.MotzerR.J., EscudierB, McDermott D.F., George S., HammersH.J., SrinivasS., TykodiS.S., Sosman J.A., ProcopioG., PlimackE.R., CastellanoD., Choueiri T.K., Gurney H., Donskov F., Bono P., Wagstaff J., Gauler T.C., Ueda T., Tomita Y., Schutz F.A., Kollmannsberger C., Larkin J., Ravaud A., Simon J.S., Xu L.A., Waxman I.M., Sharma P. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015; 373(19): 1803-13. doi: 10.1056/NEJMoa1510665.

10. Bristol-Myers Squibb (2014) Phase 2 objective response rate and survival data for Opdivo (nivolumab) in heavily pre-treated advanced squamous cell non-small cell lung cancer. To be presented at the 2014 Chicago multidisciplinary symposium on thoracic oncology [Internet]. Bristol-Myers Squibb Company; 2014. URL: https://news.bms.com/news/ details/2014/Phase-2-0bjective-Response-Rate-and-Survival-Data-for-Opdivo-nivolumab-in-Heavily-Pre-treated-Advanced-Squamous-Cell-Non-Small-Cell-Lung-Cancer-to-be-Presented-at-the-2014-Chicago-Multidisciplinary-Symposium-on-Thoracic-Oncology/default.aspx. [cited 2022 June 25].

11. OvermanM.J., McDermott R., Leach J.L., Lonardi S., Lenz H.J., Morse M.A., Desai J., Hill A., Axelson M., Moss R.A., Goldberg M.V, Cao Z.A., Ledeine J.M., Maglinte G.A., Kopetz S., Andre T. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 2017; 18(9): 1182-91. doi: 10.1016/ S1470-2045(17)30422-9.

12. Morris V.K., Salem M.E., Nimeiri H., Iqbal S., Singh P., Ciombor K., PoliteB., DemingD., ChanE., Wade J.L., XiaoL., Bekaii-Saab T., VenceL., Blando J., MahvashA., Foo W.C., Ohaji C., PasiaM., BlandG., Ohinata A., Rogers J., Mehdizadeh A., Banks K., LanmanR., Wolff R.A., Streicher H., Allison J., Sharma P., Eng C. Nivolumab for previously treated unresect-able metastatic anal cancer (NCI9673): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017; 18(4): 446-53. doi: 10.1016/S1470-2045-(17)30104-3.

13. Yamamoto N., Nokihara H., Yamada Y., Shibata T., Tamura Y., Seki Y., HondaK., Tanabe Y., WakuiH., Tamura T. Phase I study ofNivolum-ab, an anti-PD-1 antibody, in patients with malignant solid tumors. Invest New Drugs. 2017; 35(2): 207-16. doi: 10.1007/s10637-016-0411-2.

14. Brahmer J.R., Drake C.G., Wollner I., Powderly J.D., Picus J., Sharfman W.H., Stankevich E., Pons A., Salay T.M., McMiller T.L., Gil-son M.M., Wang C., SelbyM., Taube J.M., AndersR., ChenL., KormanA.J., Pardoll D.M., Lowy I., Topalian S.L. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010; 28(19): 3167-75. doi: 10.1200/JC0.2009.26.7609.

15. Iwai Y., Ishida M., Tanaka Y., Okazaki T., Honjo T., Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 2002; 99(19): 12293-7. doi: 10.1073/pnas.192461099.

16. FDA grants nivolumab accelerated approval for MSI-H or dMMR colorectal cancer [Internet]. U.S. Food and Drug Administration; 2017. URL: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ ucm569366.htm. [cited 2022 Jun 29].

17. Brahmer J.R., Tykodi S.S., Chow L.Q., Hwu W.J., Topalian S.L., HwuP., Drake C.G., CamachoL.H., Kauh J., OdunsiK., PitotH.C., Hamid O., Bhatia S., MartinsR., EatonK., Chen S., Salay T.M., Alaparthy S., Grosso J.F., Korman A.J., Parker S.M., Agrawal S., Goldberg S.M., Par-doll D.M., Gupta A., Wigginton J.M. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012; 366(26): 2455-65. doi: 10.1056/NEJMoa1200694.

18. Mollica V., Di Nunno V, Corcioni B., Fiorentino M., Nobili E., SchiavinaR., GolfieriR., BrunocillaE., ArdizzoniA., MassariF. A case of complete response to nivolumab after long-term progression-free survival with tyrosine kinase inhibitor. Anticancer Drugs. 2018; 29(9): 911-3. doi: 10.1097/CAD.0000000000000663.

19. Namikawa T., Ishida N., Tsuda S., Fujisawa K., Munekage E., Iwabu J., Munekage M., Uemura S., Tsujii S., Maeda H., Kitagawa H., KobayashiM., Hanazaki K. Successful treatment of liver metastases arising from early gastric cancer achieved clinical complete response by nivolum-ab. Surg Case Rep. 2018; 4(1): 71. doi: 10.1186/s40792-018-0479-3.

20. Borcoman E., Kanjanapan Y., Champiat S., Kato S., Servois V, Kurzrock R., Goel S., Bedard P., Tourneau C.Le. Novel patterns of response under immunotherapy. Ann Oncol. 2019; 30(3): 385-96. doi: 10.1093/ annonc/mdz003.

21. Tazdait M., MezquitaL., Lahmar J., FerraraR., BidaultF., Amma-ri S., Balleyguier C., PlanchardD., Gazzah A., Soria G.C., Marabelle A., BesseB., Caramella C. Patterns of responses in metastatic NSCLC during PD-1 or PDL-1 inhibitor therapy: comparison of RECIST 1.1, irRECIST and iRECIST criteria. Eur J Cancer. 2018; 88: 38-47. doi: 10.1016/j. ejca.2017.10.017.

22. Lipson E.J., Sharfman W.H., Drake C.G., Wollner I., Taube J.M., Anders R.A., Xu H., Yao S., PonsA., Chen L., Pardoll D.M., Brahmer J.R., Topalian S.L. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res. 2013; 19(2): 462-8. doi: 10.1158/1078-0432.CCR-12-2625.

23. Ruiz-Banobre J., Anido U., Abdulkader I., Antunez-Lopez J., LopezLopez R., Garcia-Gonzalez J. Long-term response to nivolumab and acute renal failure in a patient with metastatic papillary renal cell carcinoma and a PD-L1 tumor expression increased with sunitinib therapy: a case report. Front Oncol. 2016; 6: 250. doi: 10.3389/fonc.2016.00250.

24. Locati L.D., Serafini M.S., Carenzo A., Canevari S., Perrone F., Orlandi E., Delbue S., Cavalieri S., Berzeri G., PichiecchioA., LicitraL.F., Marchioni E., De Cecco L. Complete response to nivolumab in recurrent/ metastatic HPV-positive head and neck squamous cell carcinoma patient after progressive multifocal leukoencephalopathy: a case report. Front Oncol. 2022; 11: 799453. doi: 10.3389/fonc.2021.799453.

25. OpreaA., BenasA., HavasiA., GorzoA. Long term administration of nivolumab for metastatic melanoma: a case report. Cureus. 2022; 14(6). doi: 10.7759/cureus.26359.

26. Wolchok J.D., Hamid O., Ribas A., Robert C., Kefford R., Hwu W-J. Weber J.S., JoshuaA.M., Gangadhar T.C., DroncaR.S., Daud A., Patnaik A., Joseph R.W., Zarour M.H., Li N.X., Xue D., Ebbinghaus S., Kang S.P., Perrone A.M., Hodi F.S. A typical patterns of response in patients (pts) with metastatic melanoma treated with pembrolizumab (MK-3475) in KEYNOTE-001. J Clin Oncol. 2015; 33(15): 3000. doi: 10.1200/ jco.2015.33.15_suppl.3000.

27. Ferris R.L., Blumenschein G.Jr., Fayette J., Guigay J., Colevas A.D., Licitra L., Harrington K., Kasper S., Vokes E.E., Even C., Worden F., Saba N.F., Iglesias Docampo L.C., Haddad R., Rordorf T., Kiyota N., Tahara M., Monga M., Lynch M., Geese W.J., Kopit J., Shaw J.W., Gil-lison M.L. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016; 375(19): 1856-67. doi: 10.1056/ NEJMoa1602252.

28. Parseghian C.M., Patnana M., Bhosale P., Hess K.R., Shih Y.T., Kim B., Kopetz S., Overman M.J., Varadhachary G.R., Javle M., Na-

ing A., Piha-Paul S., Hong D., Le H., Subbiah V., Pant S. Evaluating for pseudoprogression in colorectal and pancreatic tumors treated with immunotherapy. J Immunother. 2018; 41(6): 284-91. doi: 10.1097/ CJI.0000000000000222.

29. Chae Y.K., Wang S., Nimeiri H., Kalyan A., Giles F.J. Pseudoprogression in microsatellite instability-high colorectal cancer during treatment with combination T cell mediated immunotherapy: a case report and literature review. Oncotarget. 2017; 8(34): 57889-97. doi: 10.18632/ oncotarget.18361.

30. Colle R., Radzik A., Cohen R., Pellat A., Lopez-Tabada D., Cachanado M., Duval A., Svrcek M., Menu Y., Andre T. Pseudoprogression in patients treated with immune checkpoint inhibitors for microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Eur J Cancer. 2021; 144: 9-16. doi: 10.1016/j.ejca.2020.11.009.

31. Schuler A., Omlin A., HusarikD.B., DeneckeB., Rothermundt C. Hyper- or pseudoprogression? A case report of MSI-high metastatic colorectal cancer. Ann Clin Case Rep. 2021; 6.

32. Guarini C., Todisco A., Tucci M., Porta C., Mannavola F. Massive hyper-progression during anti-PD-1 immunotherapy in a young patient with metastatic mucinous adenocarcinoma of the right colon: a case report and literature review. Precis Cancer Med. 2021; 4: 30. doi: 10.21037/pcm-21-10.

33. Wang Z., Liu C., Bai Y., Zhao X., Cui L., Peng Z., Zhang X., Wang X., Zhao Z., Li J., Shen L. Redefine hyperprogressive disease during treatment with immune-checkpoint inhibitors in patients with gastrointestinal cancer. Front. Oncol. 2021; 11: 761110. doi: 10.3389/fonc.2021.761110.

34. ChanK.H., Lakkasani S., Ramahi A., ShaabanH.S. Hyperprogressive disease in an advanced stage colon cancer patient on pembrolizumab: a case report. Cureus. 2020; 12(4). doi: 10.7759/cureus.7764.

35. Zhou W., Zhou Y., Yi C., ShuX., Wei G., ChenX., ShenX., QiuM. Case report: immune and genomic characteristics associated with hyperprogression in a patient with metastatic deficient mismatch repair gastrointestinal cancer treated with anti-PD-1 antibody. Front. Immunol. 2021; 12: 749204. doi: 10.3389/fimmu.2021.749204.

36. Napolitano M., Trudu L., Bertolini F. Unexpected response to nivolumab in a "Fast Progressor" head and neck cancer patient. Case Rep Oncol. 2019; 12(3): 709-14. doi: 10.1159/000502858.

37. Tozuka T., Kitazono S., Sakamoto H., Yoshida H., Amino Y., Uematsu S., Yoshizawa T., Hasegawa T., Uchibori K., Yanagitani N., Horiike A., Horai T., Seike M., Gemma M., Nishio M. Dissociated responses at initial computed tomography evaluation is a good prognostic factor in non-small cell lung cancer patients treated with anti-programmed cell death-1/ligand 1 inhibitors. BMC Cancer. 2020; 20(1): 207. doi: 10.1186/ s12885-020-6704-z.

38. Humbert O., Cadour N., Paquet M., Schiappa R., Poudenx M., Chardin D., Borchiellini D., Benisvy D., Ouvrier M.J., Zwarthoed C., Schiazza A., Ilie M., Ghalloussi H., Koulibaly P.M., Darcourt J., Otto J. 18FDG PET/CT in the early assessment of non-small cell lung cancer response to immunotherapy: frequency and clinical significance of atypical evolutive patterns. Eur J Nucl Med Mol Imaging. 2020; 47(5): 1158-67. doi: 10.1007/s00259-019-04573-4.

39. de Velasco G., Krajewski K.M., Albiges L., AwadM.M., Bellmunt J., Hodi F.S., Choueiri T.K. Radiologic heterogeneity in responses to anti-PD-1/PD-L1 therapy in metastatic renal cell carcinoma. Cancer Immunol Res. 2016; 4(1): 12-7. doi: 10.1158/2326-6066.CIR-15-0197.

40. Wolchok J.D., HoosA., O'Day S., Weber J.S., Hamid O., Lebbé C., Maio M., Binder M., Bohnsack O., Nichol G., Humphrey R., Hodi F.S. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009; 15(23): 7412-20. doi: 10.1158/1078-0432.CCR-09-1624.

41. NishinoM., Giobbie-Hurder A., GarganoM., SudaM., RamaiyaN.H., Hodi F.S. Developing a common language for tumor response to immuno-therapy: immune-related response criteria using unidimensional measurements. Clin Cancer Res. 2013; 19(14): 3936-43. doi: 10.1158/1078-0432. CCR-13-0895.

42. Seymour L., Bogaerts J., Perrone A., Ford R., Schwartz L.H., Man-drekar S., Lin N.U., Litière S., Dancey J., Chen A., Hodi F.S., Therasse P., Hoekstra O.S., Shankar L.K., Wolchok J.D., Ballinger M., Caramella C., de Vries E.G.E.; RECIST working group. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017; 18(3): 143-52. doi: 10.1016/S1470-2045(17)30074-8. Erratum in: Lancet Oncol. 2019; 20(5): 242.

43. Hodi F.S., Ballinger M., Lyons B., Soria J.C., Nishino M., Tabernero J., Powles T., Smith D., Hoos A., McKenna C., Beyer U., Rhee I., Fine G., Winslow N., Chen D.S., Wolchok J.D. Immune-Modified Response Evaluation Criteria In Solid Tumors (imRECIST): Refining Guidelines to Assess the Clinical Benefit of Cancer Immunotherapy. J Clin Oncol. 2018; 36(9): 850-8. doi: 10.1200/Jœ.2017.75.1644.

Поступила/Received 30.08.2022 Одобрена после рецензирования/Revised 20.10.2022 Принята к публикации/Accepted 25.10.2022

about the authors

Nataliya N. Babyshkina, MD, DSc, Senior Researcher, Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences; Associate Professor of the Department of Biochemistry and Molecular Biology with a course in clinical laboratory diagnostics, Siberian State Medical University of the Ministry of Health of Russia (Tomsk, Russia). E-mail: nbabyshkina@mail.ru. Author ID (Scopus): 26641099700. Researcher ID (WOS): A-7526-2012. ORCID: 0000-0002-0562-3878.

Tatyana A. Dronova, MD, Junior Researcher, Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Author ID (Scopus): 6602971247. Researcher ID (WOS): R-5952-2016. ORCID: 0000-0003-3009-2404.

Polina A. Gervas, MD, PhD, Researcher, Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Author ID (Scopus): 13613767400. Researcher ID (WOS): C-5846-2012. ORCID: 0000-0003-0051-8814.

Nataliya O. Popova, MD, PhD, Senior Researcher, Chemotherapy Department, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Author ID (Scopus): 7201879486. Researcher ID (WOS): I-9417-2017. ORCID: 0000-0001-5294-778X.

Alexey Yu. Dobrodeev, MD, DSc, Leading Researcher, Department of Abdominal Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Author ID (Scopus): 24832974200. Researcher ID (WOS): B-5644-2017. ORCID: 0000-0002-2748-0644.

Dmitry N. Kostromitsky, MD, PhD, Researcher, Department of Abdominal Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). ORCID: 0000-0001-5691-2349.

Sergey G. Afanasyev, MD, DSc, Professor, Head of the Department of Abdominal Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Researcher ID (WOS): D-2084-2012. Author ID (Scopus): 7005336732. ORCID: 0000-0002-4701-0375.

Viktor E. Goldberg, MD, DSc, Professor, Head of Chemotherapy Department, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Author ID (Scopus): 7005773126. Researcher ID (WOS): C-8911-2012. ORCID: 0000-0003-4753-5283.

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

Nadezhda V. Cherdyntseva, DSc, Professor, Corresponding Member of the Russian Academy of Sciences, Professor, Head of the Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences (Tomsk, Russia). Author ID (Scopus): 6603911744. Researcher ID (WOS): C-7943-2012. ORCID: 0000-00031526-901.

author contribution

Nataliya N. Babyshkina: study conception, literature review, writing of the manuscript. Tatyana A. Dronova: data analysis, drafting of the manuscript. Polina A. Gervas: data analysis, drafting of the manuscript.

Nataliya O. Popova: critical review and introduction of valuable intellectual content, approval of the final version of the manuscript.

Alexey Yu. Dobrodeev: data analysis, drafting of the manuscript.

Dmitry N. Kostromitsky: data analysis, drafting of the manuscript.

Sergey G. Afanasyev: study conception.

Viktor E. Goldberg: study conception.

Nadezhda V. Cherdyntseva: study conception.

Funding

The study was supported by the Russian Science Foundation grant No. 22-15-00212 dated May 13, 2022 "Transcriptomic and proteomic markers for the prognosis and effectiveness of therapy for metastatic colon cancer".

Conflict of interests

The authors declare that they have no conflict of interest.

СВЕДЕНИЯ ОБ АВТОРАХ

Бабышкина Наталия Николаевна, доктор медицинских наук, старший научный сотрудник лаборатории молекулярной онкологии и иммунологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук; доцент кафедры биохимии и молекулярной биологии с курсом клинической лабораторной диагностики, ФГБОУ ВО «Сибирский государственный медицинский университет» Минздрава России (г. Томск, Россия). E-mail: nbabyshkina@mail.ru. SPIN-код: 2738-9275. Author ID (Scopus): 26641099700. Researcher ID (WOS): A-7526-2012. ORCID: 0000-0002-0562-3878.

Дронова Татьяна Анатольевна, младший научный сотрудник лаборатории молекулярной онкологии и иммунологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 3516-2517. Author ID (Scopus): 6602971247. Researcher ID (WOS): R-5952-2016. ORCID: 0000-0003-3009-2404.

Гервас Полина Анатольевна, кандидат медицинских наук, научный сотрудник лаборатории молекулярной онкологии и иммунологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 2934-7970. Author ID (Scopus): 13613767400. Researcher ID (WOS): C-5846-2012. ORCID: 0000-0003-0051-8814.

Попова Наталия Олеговна, кандидат медицинских наук, старший научный сотрудник отделения химиотерапии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 7672-1029. Author ID (Scopus): 7201879486. Researcher ID (WOS): I-9417-2017. ORCID: 0000-0001-5294-778X.

Добродеев Алексей Юрьевич, доктор медицинских наук, ведущий научный сотрудник отделения абдоминальной онкологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 5510-4043. Author ID (Scopus): 24832974200. Researcher ID (WOS): B-5644-2017. ORCID: 0000-0002-2748-0644.

Костромицкий Дмитрий Николаевич, кандидат медицинских наук, научный сотрудник отделения абдоминальной онкологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 9466-6641. ORCID: 0000-0001-5691-2349.

Афанасьев Сергей Геннадьевич, доктор медицинских наук, профессор, заведующий отделением абдоминальной онкологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 9206-3037. Researcher ID (WOS): D-2084-2012. Author ID (Scopus): 7005336732. ORCID: 0000-0002-4701-0375.

Гольдберг Виктор Евгеньевич, доктор медицинских наук, профессор, заведующий отделением химиотерапии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 7587-0560. Author ID (Scopus): 7005773126. Researcher ID (WOS): C-8911-2012. ORCID: 0000-0003-4753-5283.

Чердынцева Надежда Викторовна, доктор биологических наук, профессор, член-корр. РАН, заведующая лабораторией молекулярной онкологии и иммунологии, Научно-исследовательский институт онкологии, Томский национальный исследовательский медицинский центр Российской академии наук (г. Томск, Россия). SPIN-код: 5344-0990. Author ID (Scopus): 6603911744. Researcher ID (WOS): C-7943-2012. ORCID: 0000-0003-1526-9013.

ВКЛАД АВТОРОВ

Бабышкина Наталия Николаевна: разработка концепции научной работы, поиск и анализ современных литературных данных, написание статьи.

Дронова Татьяна Анатольевна: поиск и анализ современных литературных данных, составление черновика статьи. Гервас Полина Анатольевна: поиск и анализ современных литературных данных, составление черновика статьи. Попова Наталия Олеговна: критический пересмотр с внесением ценного интеллектуального содержания, утверждение окончательной версии статьи.

Добродеев Алексей Юрьевич: поиск и анализ современных литературных данных, написание статьи.

Костромицкий Дмитрий Николаевич: поиск и анализ современных литературных данных, составление черновика статьи.

Афанасьев Сергей Геннадьевич: разработка концепции научной работы.

Гольдберг Виктор Евгеньевич: разработка концепции научной работы.

Чердынцева Надежда Викторовна: разработка концепции научной работы.

Финансирование

Работа поддержана грантом РНФ № 22-15-00212 от 13.05.2022 «Транскриптомные и протеомные маркеры прогноза и эффективности терапии метастатического рака толстой кишки». Конфликт интересов

Авторы заявляют об отсутствии конфликта интересов.

i Надоели баннеры? Вы всегда можете отключить рекламу.