Clinicopathological variables.

Clinicopathological variables.

Source publication
Article
Full-text available
Patients with colorectal liver metastases (CLM) commonly receive neoadjuvant chemotherapy (NACT) prior to surgical resection. NACT may induce immunogenic cell death with subsequent recruitment of T-cells to the tumor microenvironment, which could be exploited by immune checkpoint inhibition (ICI). In theory, this could expand the use of ICI to obta...

Contexts in source publication

Context 1
... carcinoembryonic antigen (CEA) prior to CLM resection was 5 µg/L (IQR 3-9). The primary colorectal cancers (pCRC) included 68 (74%) located in the left colon and rectum, mostly T3 and T4 tumors (n = 86; 93%), with more than half of the cases exhibiting lymph node metastases (n = 52; 56%) ( Table 1). In univariable analysis, a CEA value above the median was associated with lower T tot values in the IT and IM regions (Supplementary Table 1). ...
Context 2
... primary colorectal cancers (pCRC) included 68 (74%) located in the left colon and rectum, mostly T3 and T4 tumors (n = 86; 93%), with more than half of the cases exhibiting lymph node metastases (n = 52; 56%) ( Table 1). In univariable analysis, a CEA value above the median was associated with lower T tot values in the IT and IM regions (Supplementary Table 1). ...
Context 3
... analyzed as a dichotomous parameter (NACT versus no-NACT), NACT administration was not associated with changes in T-cell densities (Supplementary Table 1), but the variation of T-cell densities within the NACT group was substantial, and parameters related to NACT administration were therefore explored. The time interval between NACT completion and CLM resection (resection interval) within the NACT group varied considerably (3-38 weeks). ...
Context 4
... univariable analysis, a short resection interval (using the median of 8-week as a cutoff) was strongly associated with a high T tot density in the IT with no difference found in the IM or N Li regions. The number of administered NACT cycles, the choice of NACT regimen, and radiologically assessed response to NACT were not associated with T-cell densities (Supplementary Table 1). In multivariable binary logistic regression analysis including resection interval, age, gender and CEA, only a short resection interval was associated with an increased likelihood of a T-cell density above the cohort median compared to a long resection interval (odds ratio 4.78 (95% CI 1. 19-19.19), ...
Context 5
... of the 92 patients (49%) received NACT, which was completed median 8 weeks (range 3-38) prior to CLM resection, with a median of 4 cycles administered (range 2-12 cycles) (Table 1). Fourteen patients out of the 47 who did not receive NACT were treated with chemotherapy after resection of the primary tumor but prior to CLM diagnosis, completed median 70 weeks (range 18-191 weeks) before CLM resection. ...
Context 6
... timing of surgery after NACT, the type of NACT regimen and the number of NACT cycles were decided by the multidisciplinary team and these parameters were not predefined by the OSLO-COMET study protocol. 27 All except one patient (who had oxaliplatin monotherapy) received fluoropyrimidine-based treatment with addition of oxaliplatin (n = 31, 1 with addition of epidermal growth factor receptor antibody), irinotecan (n = 6; 3 in combination with bevacizumab, 2 with epidermal growth factor receptor antibody), or were converted from oxaliplatin to irinotecan (because of toxicity or poor response; n = 3) ( Table 1). ...

Similar publications

Article
Full-text available
Currently, there is a huge demand to develop chemoimmunotherapy with reduced systemic toxicity and potent efficacy to combat late-stage cancers with spreading metastases. Here, we report several “cocktail” therapeutic formulations by mixing immunogenic cell death (ICD)–inducing chemotherapeutics and immune adjuvants together with alginate (ALG) for...

Citations

... 21 It was recently shown that the abundance and spatial distribution of different immune-cell subsets within MSS-CRC metastases exposed to neoadjuvant chemotherapy (treatment duration not reported) varied in an organ-specific manner; in liver metastases, activated T cells were enriched in the outer invasive margin, but not in the tumor core as seen in lung metastases and primary tumors. 22 Yet, we previously showed that neoadjuvant chemotherapy caused a transient increase in intratumoral T-cell density of MSS-CRC liver metastases, 23 a response retrieved in the case reported here by the expanded clonal TCR repertoires of the primarily relapsed liver metastases, lost shortly thereafter by the end-stage metastases. ...
Article
Full-text available
The randomized METIMMOX trial (NCT03388190) examined if patients with previously untreated, unresectable abdominal metastases from microsatellite-stable (MSS) colorectal cancer (CRC) might benefit from potentially immunogenic, short-course oxaliplatin-based chemotherapy alternating with immune checkpoint blockade (ICB). Three of 38 patients assigned to this experimental treatment had metastases from BRAF-mutant MSS-CRC, in general a poor-prognostic subgroup explored here. The ≥70-year-old females presented with ascending colon adenocarcinomas with intermediate tumor mutational burden (6.2–11.8 mutations per megabase). All experienced early disappearance of the primary tumor followed by complete response of all overt metastatic disease, resulting in progression-free survival as long as 20–35 months. However, they encountered recurrence at previously unaffected sites and ultimately sanctuary organs, or as intrahepatic tumor evolution reflected in the terminal loss of initially induced T-cell clonality in liver metastases. Yet, the remarkable first-line responses to short-course oxaliplatin-based chemotherapy alternating with ICB may offer a novel therapeutic option to a particularly hard-to-treat MSS-CRC subgroup.
... Colorectal cancer (CRC) is the fourth cause of cancer deaths worldwide [1], with metastasis the critical cause of CRC-related death [13]. CRLM represents the distant metastatic disease for 50% of CRC patients [19] occurring as synchronous or after primary tumor removal. A small subset of CRC cells evade from the primary CRC undergoing morphological changes such as epithelial-to-mesenchymal transition (EMT), migration through the extracellular matrix (ECM) and invasion into the neighboring tissues [13]. ...
Article
Full-text available
Background and purpose While HCC is an inflammation-associated cancer, CRLM develops on permissive healthy liver microenvironment. To evaluate the immune aspects of these two different environments, peripheral blood-(PB), peritumoral-(PT) and tumoral tissues-(TT) from HCC and CRLM patients were evaluated. Methods 40 HCC and 34 CRLM were enrolled and freshly TT, PT and PB were collected at the surgery. PB-, PT- and TT-derived CD4 ⁺ CD25 ⁺ Tregs, M/PMN-MDSC and PB-derived CD4 ⁺ CD25 ⁻ T-effector cells (Teffs) were isolated and characterized. Tregs’ function was also evaluated in the presence of the CXCR4 inhibitor, peptide-R29, AMD3100 or anti-PD1. RNA was extracted from PB/PT/TT tissues and tested for FOXP3, CXCL12, CXCR4, CCL5, IL-15, CXCL5, Arg-1, N-cad, Vim, CXCL8, TGFβ and VEGF-A expression. Results In HCC/CRLM-PB, higher number of functional Tregs, CD4 ⁺ CD25 hi FOXP3 ⁺ was detected, although PB-HCC Tregs exert a more suppressive function as compared to CRLM Tregs. In HCC/CRLM-TT, Tregs were highly represented with activated/ENTPD-1 ⁺ Tregs prevalent in HCC. As compared to CRLM, HCC overexpressed CXCR4 and N-cadherin/vimentin in a contest rich in arginase and CCL5. Monocytic MDSCs were highly represented in HCC/CRLM, while high polymorphonuclear MDSCs were detected only in HCC. Interestingly, the function of CXCR4-PB-Tregs was impaired in HCC/CRLM by the CXCR4 inhibitor R29. Conclusion In HCC and CRLM, peripheral blood, peritumoral and tumoral tissues Tregs are highly represented and functional. Nevertheless, HCC displays a more immunosuppressive TME due to Tregs, MDSCs, intrinsic tumor features (CXCR4, CCL5, arginase) and the contest in which it develops. As CXCR4 is overexpressed in HCC/CRLM tumor/TME cells, CXCR4 inhibitors may be considered for double hit therapy in liver cancer patients.
... At present, it is believed that microsatellite instability (MSI) or mismatch repair (MMR), tumor mutational burden (TMB) and the number of tumor-infiltrating T-cells have been established as a predictive biomarker for response to ICIs therapy, regardless of the primary site [28,29]. CRC can be classified into two types, MSI-H/dMMR accounting for approximately 15% of all CRCs and MSS/pMMR accounting for approximately 85% of CRCs, according to its status of MSI or MMR in a tumor [30]. ...
Article
Colorectal cancer (CRC) is one of the high incident and lethal malignant tumors, and most of the patients are diagnosed at an advanced stage. The treatment of CRC mainly includes surgery, chemotherapy, radiotherapy and molecular targeted therapy. Despite these approaches have increased overall survival (OS) of CRC patients, the prognosis of advanced CRC remains poor. In recent years, remarkable breakthroughs have been made in tumor immunotherapy, especially immune checkpoint inhibitors (ICIs) therapy, bringing long-term survival benefits to tumor patients. With the increasing wealth of clinical data, ICIs have achieved significant efficacy in the treatment of high microsatellite instability/deficient mismatch repair (MSI-H/dMMR) advanced CRC, but the therapeutic effects of ICIs on microsatellite stable (MSS) advanced CRC patients is currently unsatisfactory. As increasing numbers of large clinical trials are performed globally, patients treated with ICIs therapy also have immunotherapy-related adverse events and treatment resistance. Therefore, a large number of clinical trials are still needed to evaluate the therapeutic effect and safety of ICIs therapy in advanced CRC. This article will focus on the current research status of ICIs in advanced CRC and discuss the current predicament of ICIs treatment.
... Colorectal cancer (CRC) is the fourth cause of cancer deaths worldwide [1] with metastasis the critical cause of CRC-related death. [17] CRLM represents the distant metastatic disease for 50% of CRC patients [24] occurring as synchronous or after primary tumor removal. A small subset of CRC cells acquires a capacity to evade from the primary CRC, through morphological changes such as epithelial-tomesenchymal transition (EMT), migration through the extracellular matrix (ECM), and invasion into the neighboring tissues. ...
Preprint
Full-text available
Background and purpose: HCC is inflammation-associated cancer and develops on chronic inflamed liver while CRLM develop on permissive healthy liver microenvironment. To evaluate the immune aspects of these two different environments, peripheral blood-(PB), peritumoral-(PT) and tumoral tissues-(TT) from HCC and CRLM patients were evaluated. Methods: 40 HCC and 34 CRLM were enrolled and freshly TT, PT and PB were collected at the surgery. PB-, PT- and TT-derived CD4⁺CD25+ Tregs and PB-derived CD4⁺CD25− Teffector cells (Teffs) were isolated and characterized for phenotype and function. Tregs function was evaluated in the presence of Peptide-R29, AMD3100 or anti-PD-1. RNA was extracted from PB/PT/TT-tissues and tested for FOXP3, CXCL12, CXCR4, CCL5, IL-15, CXCL5, Arg-1, N-cad, Vim, CXCL8, TGFb and VEGF-A expression. Results: In HCC/CRLM-PB higher number of functional Tregs, CD4⁺CD25hiFOXP3⁺ were detected, although PB-HCC Tregs exert a more suppressive function as compared to CRLM-Tregs. In HCC/CRLM-TT Tregs were highly represented with Activated/ENTPD-1⁺Tregs prevalent in HCC. As compared to CRLM, HCC overexpressed CXCR4 and N-cadherin/Vimentin in a contest rich of arginase and CCL5. Monocytic-MDSCs were highly represented in HCC/CRLM while high Polymorphonuclear-MDSCs were detected only in HCC. Interestingly, CXCR4-PB-Tregs inhibition, through the inhibitor-R29, impaired Tregs function in HCC/CRLM. Conclusion: in HCC and CRLM, peripheral blood, peritumoral and tumoral tissues-Tregs are highly represented and functional. Nevertheless, HCC display a more immunosuppressive TME due to Tregs, MDSCs, intrinsic tumor features (CXCR4, CCL5, arginase) and the contest in which it develops. As CXCR4 is overexpressed in HCC/CRLM tumor/TME cells, CXCR4 inhibitors may be considered for double hits therapy in liver cancer patients.
... However, a follow-up study showed that there was no association between neoadjuvant chemotherapy and intratumoral T cell density within colorectal liver metastases. Investigating further, it appeared that this discrepancy may have been temporal in nature, being associated with a transient increase in T cell density, as there was a significant difference in T cell infiltrate between patients who received neoadjuvant chemotherapy fewer than 9.5 weeks before resection of liver metastases and those who did not receive chemotherapy or those who had a longer interval between treatment and resection [111]. The implications of these findings not only described possible biomarkers but also gave better comprehension that can inform optimal treatment schedules combining chemotherapy and immunotherapy. ...
Article
Full-text available
Innovative strategies to re-establish the immune-mediated destruction of malignant cells is paramount to the success of anti-cancer therapy. Accumulating evidence suggests that radiotherapy and select chemotherapeutic drugs and small molecule inhibitors induce immunogenic cell stress on tumors that results in improved immune recognition and targeting of the malignant cells. Through immunogenic cell death, which entails the release of antigens and danger signals, and immunogenic modulation, wherein the phenotype of stressed cells is altered to become more susceptible to immune attack, radiotherapies, chemotherapies, and small-molecule inhibitors exert immune-mediated anti-tumor responses. In this review, we discuss the mechanisms of immunogenic cell death and immunogenic modulation and their relevance in the anti-tumor activity of radiotherapies, chemotherapies, and small-molecule inhibitors. Our aim is to feature the immunological aspects of conventional and targeted cancer therapies and highlight how these therapies may be compatible with emerging immunotherapy approaches.
... These results suggested that NACT recruited immune cells in patients with colorectal cancer liver metastases, but the recruited CD8 + T cells might induce immune tolerance, which is responsible for the poor outcome of certain patients. Another study found time-dependent alternations in T cells, with T cells increasing in the group that underwent surgical resection only within a short interval (<9.5 weeks) after the completion of NACT (124). Moreover, the Treg/CTL ratio was lower in the short-interval group (124). ...
... Another study found time-dependent alternations in T cells, with T cells increasing in the group that underwent surgical resection only within a short interval (<9.5 weeks) after the completion of NACT (124). Moreover, the Treg/CTL ratio was lower in the short-interval group (124). This study illustrated that the immunosuppressive milieu gradually becomes prominent after NACT, while the time-dependent changes in immune cells indicated that there might be a time window of opportunity for application of immunotherapy. ...
Article
Full-text available
In recent years, numerous studies have demonstrated that the tumor immune microenvironment (TIME) is capable of regulating the growth of tumors, and tumor-infiltrating immune cells in the TIME can affect the prognosis and treatment responses of patients. Consequently, therapies targeting these immune cells have emerged as important antitumor treatments. As a crucial componet of the perioperative treatment of malignant tumors, neoadjuvant chemotherapy (NACT) can improve the surgical resection rate and prognosis of patients and is a suitable clinical model to evaluate the effect of chemotherapy on the TIME. To provide a rationale for developing valid combinational therapies, this review summarizes the impact of NACT on the TIME, the relationship between tumor-infiltrating immune cells and treatment responses of patients, and the prognostic value of these infiltrating immune cells.
... Yet, preclinical [15,16] and clinical evidence, including our own from studies applying short-course oxaliplatin-based chemotherapy in combined-modality treatment schedules [17][18][19][20][21][22], supports the notion that oxaliplatin may induce immunogenic cell death [23] in CRC and invoke efficacious antitumour immunity. This led us to hypothesise that the metastatic MSS/pMMR-CRC entity can be transformed into an immunogenic condition by two cycles of the oxaliplatin-containing Nordic FLOX regimen [24], and patients with previously untreated, the unresectable disease that predominantly comprises infradiaphragmatic manifestations may achieve therapeutic efficacy from the sequential addition of ICB therapy. ...
Article
Full-text available
Background: Immune checkpoint blockade (ICB) results in radiologic tumour response dynamics that differ from chemotherapy efficacy measures and require an early signal of clinical utility. Methods: Previously untreated, unresectable microsatellite-stable (MSS)/mismatch repair-proficient (pMMR) colorectal cancer (CRC) patients were randomly assigned to the oxaliplatin-based Nordic FLOX regimen (control arm) or repeat sequential two FLOX cycles and two ICB cycles (experimental arm). The radiologic response was assessed every 8 weeks. In this post hoc analysis, we explored early target lesion (TL) dynamics as indicator of ICB responsiveness. Progression-free survival (PFS) was the primary endpoint. Results: Using a landmark analysis approach, we categorised experimental-arm patients into ≥10% (N = 19) or <10% (N = 16) TL reduction at the first post-baseline response assessment. Median PFS for the groups was 16.0 (95% confidence interval (CI), 12.3-19.7) and 3.9 months (95% CI, 2.3-5.5), respectively, superior and inferior (both P < 0.01) to the median PFS of 9.8 months (95% CI, 4.9-14.7) for control arm patients (N = 31). Conclusions: Radiologic TL reduction of ≥10% at the first post-baseline response assessment identified patients with ICB-responsive metastatic MSS/pMMR-CRC. This pragmatic measure may be used to monitor patients in investigational ICB schedules, enabling early treatment adaptation for unresponsive cases. Trial registration: ClinicalTrials.gov number, NCT03388190 (02/01/2018).
... The tumor microenvironment is a complex and dynamic system that contains a variety of immune cells. Tumor-infiltrating T lymphocytes are regarded as the main effectors of anti-tumor immune response [8,9], and CD8 is recognized as the important marker of T-cell infiltration. Foxp3 is an important marker of regulatory T cells. ...
Article
Full-text available
Background: Liver metastases are a major contributor to the poor immunotherapy response in colorectal cancer patients. However, the distinctions in the immune microenvironment between primary tumors and liver metastases are poorly characterized. The goal of this study was to compare the expression profile of multiple immune cells to further analyze the similarities and differences between the microenvironments of liver metastases and the primary tumor. Methods: Tissues from 17 patients with colorectal cancer who underwent resection of primary and liver metastases was analyzed using multispectral immunofluorescence. The expression of multiple immune cells (CD8, Foxp3, CD68, CD163, CD20, CD11c, CD66b, CD56, PD-L1, INF-γ, Ki67 and VEGFR-2) in the tumor center (TC), tumor invasive front (< 150 µm from the tumor center, TF) and peritumoral region (≥ 150 µm from the tumor center, PT) was evaluated via comparison. The expression of CD68 and CD163 in different regions was further analyzed based on the cell colocalization method. In addition, different immune phenotypes were studied and compared according to the degree of CD8 infiltration. Results: The expression trends of 12 markers in the TF and TC regions were basically the same in the primary tumor and liver metastasis lesions. However, in comparison of the TF and PT regions, the expression trends were not identical between primary and liver metastases, especially CD163, which was more highly expressed in the PT region relative to the TF region. In the contrast of different space distribution, the expression of CD163 was higher in liver metastases than in the primary foci. Further analysis of CD68 and CD163 via colocalization revealed that the distribution of macrophages in liver metastases was significantly different from that in the primary foci, with CD68-CD163+ macrophages predominating in liver metastases. In addition, among the three immunophenotypes, CD163 expression was highest in the immune rejection phenotype. Conclusions: The immune cells found in the primary tumors of colorectal cancer differed from those in liver metastases in terms of their spatial distribution. More immunosuppressive cells were present in the liver metastases, with the most pronounced differential distribution found for macrophages. CD68-CD163+ macrophages may be associated with intrahepatic immunosuppression and weak immunotherapeutic effects.
... The tumor microenvironment is a complex and dynamic system that contains a variety of immune cells. Tumor-infiltrating T lymphocytes are regarded as the main effectors of antitumor immune response (8,9), and CD8 is recognized as the important marker of T-cell infiltration. Foxp3 is an important marker of regulatory T cells. ...
... In this study, we used PerkinElmer Inform software to compare and analyze various aspects of primary colorectal cancer and liver metastases (T cells, B cells, macrophages, neutrophils, DCs, NK cells, lymphokines, PD-L1, tumor angiogenesis and cell proliferation). Previous research has mainly focused on T lymphocyte (8,31), but T cells are insufficient for representing the complex microenvironment of the immune system. Macrophages, B cells, neutrophils, etc, also play an important role in the tumor microenvironment (32,33). ...
Preprint
Full-text available
Background Liver metastases are a major contributor to the poor immunotherapy response in colorectal cancer patients. However, the distinctions in the immune microenvironment between primary tumors and liver metastases are poorly characterized. The goal of this study was to compare the expression profile of multiple immune cells to further analyze the similarities and differences between the microenvironments of liver metastases and the primary tumor. Methods Tissues from 17 patients with colorectal cancer who underwent resection of primary and liver metastases was analyzed using multispectral immunofluorescence. The expression of multiple immune cells (CD8, Foxp3, CD68, CD163, CD20, CD11c, CD66b, CD56, PD-L1, INF-γ, Ki67 and VEGFR-2) in the tumor center (TC), tumor invasive front (<150 µm from the tumor center, TF) and peritumoral region (≥150 µm from the tumor center, PT) was evaluated via comparison. The expression of CD68 and CD163 in different regions was further analyzed based on the cell colocalization method. In addition, different immune phenotypes were studied and compared according to the degree of CD8 infiltration. Results The expression trends of 12 markers in the TF and TC regions were basically the same in the primary tumor and liver metastasis lesions. However, in comparison of the TF and PT regions, the expression trends were not identical between primary and liver metastases, especially CD163, which was more highly expressed in the PT region relative to the TF region. In the contrast of different space distribution, the expression of CD163 was higher in liver metastases than in the primary foci. Further analysis of CD68 and CD163 via colocalization revealed that the distribution of macrophages in liver metastases was significantly different from that in the primary foci, with CD68⁻CD163⁺ macrophages predominating in liver metastases. In addition, among the three immunophenotypes, CD163 expression was highest in the immune rejection phenotype. Conclusions The immune cells found in the primary tumors of colorectal cancer differed from those in liver metastases in terms of their spatial distribution. More immunosuppressive cells were present in the liver metastases, with the most pronounced differential distribution found for macrophages. CD68⁻CD163⁺ macrophages may be associated with intrahepatic immunosuppression and weak immunotherapeutic effects.
... In a further study investigating this patient population, it was shown that while there was no association between neoadjuvant chemotherapy and intratumoral T-cell density within colorectal liver metastases, there was a significant increase in intratumoral T-cell density in patients who received neoadjuvant chemotherapy fewer than 9.5 weeks before liver metastases resection compared to both patients with a longer interval and those who did not receive chemotherapy. This result is highly interesting and could be an important data point when designing clinical strategies meant to take advantage of chemotherapy-induced immunogenic cell death (113). ...
Article
Full-text available
Cancer treatment has rapidly entered the age of immunotherapy, and it is becoming clear that the effective therapy of established tumors necessitates rational multi-combination immunotherapy strategies. But even in the advent of immunotherapy, the clinical role of standard-of-care chemotherapy regimens still remains significant and may be complementary to emerging immunotherapeutic approaches. Depending on dose, schedule, and agent, chemotherapy can induce immunogenic cell death, resulting in the release of tumor antigens to stimulate an immune response, or immunogenic modulation, sensitizing surviving tumor cells to immune cell killing. While these have been previously defined as distinct processes, in this review we examine the published mechanisms supporting both immunogenic cell death and immunogenic modulation and propose they be reclassified as similar effects termed “immunogenic cell stress.” Treatment-induced immunogenic cell stress is an important result of cytotoxic chemotherapy and future research should consider immunogenic cell stress as a whole rather than just immunogenic cell death or immunogenic modulation. Cancer treatment strategies should be designed specifically to take advantage of these effects in combination immunotherapy, and novel chemotherapy regimens should be designed and investigated to potentially induce all aspects of immunogenic cell stress.