Upfront dose-reduced chemotherapy synergizes with immunotherapy to optimize chemoimmunotherapy in squamous cell lung carcinoma

Xiran He, Yang Du, Zhijie Wang, Xin Wang, Jianchun Duan, Rui Wan, Jiachen Xu, Pei Zhang, Di Wang, Yanhua Tian, Jiefei Han, Kailun Fei, Hua Bai, Jie Tian, Jie Wang, Xiran He, Yang Du, Zhijie Wang, Xin Wang, Jianchun Duan, Rui Wan, Jiachen Xu, Pei Zhang, Di Wang, Yanhua Tian, Jiefei Han, Kailun Fei, Hua Bai, Jie Tian, Jie Wang

Abstract

Background: The survival benefits of combining chemotherapy (at the maximum tolerated dose, MTD) with concurrent immunotherapy, collectively referred to as chemoimmunotherapy, for the treatment of squamous cell lung carcinoma (SQCLC) have been confirmed in recent clinical trials. Nevertheless, optimization of chemoimmunotherapy in order to enhance the efficacy of immune checkpoint inhibitors (ICIs) in SQCLC remains to be explored.

Methods: Cell lines, syngeneic immunocompetent mouse models, and patients' peripheral blood mononuclear cells were used in order to comprehensively explore how to enhance ectopic lymphoid-like structures (ELSs) and upregulate the therapeutic targets of anti-programmed death 1 (PD-1)/anti-PD-1 ligand (PD-L1) monoclonal antibodies (mAbs), thus rendering SQCLC more sensitive to ICIs. In addition, molecular mechanisms underlying optimization were characterized.

Results: Low-dose chemotherapy contributed to an enhanced antigen exposure via the phosphatidylinositol 3-kinase/Akt/transcription factor nuclear factor kappa B signaling pathway. Improved antigen uptake and presentation by activated dendritic cells (DCs) was observed, thus invoking specific T cell responses leading to systemic immune responses and immunological memory. In turn, enhanced antitumor ELSs and PD-1/PD-L1 expression was observed in vivo. Moreover, upfront metronomic (low-dose and frequent administration) chemotherapy extended the time window of the immunostimulatory effect and effectively synergized with anti-PD-1/PD-L1 mAbs. A possible mechanism underlying this synergy is the increase of activated type I macrophages, DCs, and cytotoxic CD8+ T cells, as well as the maintenance of intestinal gut microbiota diversity and composition. In contrast, when combining routine MTD chemotherapy with ICIs, the effects appeared to be additive rather than synergistic.

Conclusions: We first attempted to optimize chemoimmunotherapy for SQCLC by investigating different combinatorial modes. Compared with the MTD chemotherapy used in current clinical practice, upfront metronomic chemotherapy performed better with subsequent anti-PD-1/PD-L1 mAb treatment. This combination approach is worth investigating in other types of tumors, followed by translation into the clinic in the future.

Keywords: combination; drug therapy; immunotherapy; lung neoplasms; lymphocytes; translational medical research; tumor-infiltrating.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Low-dose chemotherapy may induce ICD via the PI3K/Akt/NF-κB signaling pathway in SQCLC cell lines. (A and B) RNA sequencing analysis to evaluate immune-related gene expression following low-dose chemotherapeutic treatment (CDDP 4 µM, GEM 20 µM, PTX 1 µM, DTX 1 µM) for 24 hours. (C–F) Percentage of SQCLC cells expressing cell surface PD-L1, as revealed by FACS, after incubation with the indicated chemotherapy agents or controls (negative and positive) for 24 hours, with or without pretreatment with 1–3 µM BAY 11–7082 (BAY), an NF-κB inhibitor, for 30 min–3 hours. (G–I) Percentage of (G) H226, (H) H520, and (I) SK-MES cells expressing cytoplasmic HMGB-1 by FACS, after incubation with the indicated chemotherapy agents or negative control for 24 hours, with or without concurrent treatment with 0.2 µM BAY. (J–L) Percentage of (J) KLN-205, (K) H226, and (L) SK-MES cells expressing cell surface PD-L1 by FACS, after incubation with the indicated chemotherapy agents or controls for 24 hours, with or without concurrent treatment with 10 µM LY294002, a PI3K inhibitor, or 10 µM MK-2206, an Akt inhibitor. (M) Percentage of KLN-205 cells expressing cell surface MHC-class I. (N) Percentage of H226 cells expressing cell surface HLA-A/B/C, after incubation with the indicated chemotherapy agents or controls for 24 hours, with or without concurrent treatment with 10 µM LY294002 or 10 µM MK-2206. Data are presented as the mean±SD; *p

Figure 2

Low-dose chemotherapy induces HMGB-1 release…

Figure 2

Low-dose chemotherapy induces HMGB-1 release by SQCLC cells, thereby promoting DC maturation (A–D)…

Figure 2
Low-dose chemotherapy induces HMGB-1 release by SQCLC cells, thereby promoting DC maturation (A–D) and inducing systemic immune responses as well as immunological memory in vivo (E, F). KLN 205-Luc cells were incubated with 4 µM CDDP, 20 µM GEM, 1 µM DTX, or 1 µM PTX for 30 hours, after which the conditioned media were dialyzed at 4°C for 24 hours using a 10,000-MWCO dialysis tubing to remove any residual chemotherapeutic drug. Immature murine bone marrow-derived dendritic cells (BMDCs) cultured in 6-well plates (1×106 cells/mL) were then cultured in unconditioned media (negative control), media containing LPS (10 µg/mL, positive control), or different conditioned media, and were harvested 24 hours later. The maturation state of BMDCs was evaluated by measuring the cell surface co-expression of CD11c+ CD11b+ CD80+, CD11c+ CD11b+ CD86+, and CD11c+ CD11b+ MHC class II+ by FACS. (A) percentage of BMDCs expressing the above-described combinations of three different cell maturation biomarkers after the different stimuli, as revealed by FACS. (B) Proportion of total mature BMDCs in each group after the different stimuli. (C) HMGB-1 levels in the media after treatment with different chemotherapeutic agents for 30 hours, as assessed by ELISA. (D) Immature BMDCs were stimulated with 50 ng/mL or 100 ng/mL recombinant HMGB-1 for 24 hours, and FACS analysis of CD11c+ CD11b+ CD80+, CD11c+ CD11b+ CD86+, CD11c+ CD11b+ MHC class II+ expression was carried out. (E, F) Murine KLN-205-Luc cells were pretreated with 1 µM CDDP, 15 µM CDDP for 72 hours or with PBS as the negative control. These pretreated cells (1×106) were subcutaneously injected into the right flanks of mice. Splenocytes were harvested after 7 days, and IFN-γ secretion was evaluated using ELISPOT, according to the kit manufacturer’s protocol. (G) The aforementioned mice were rechallenged with 2×105 live KLN-205 cells in the opposite flank, and tumor growth was monitored using vernier calipers and BLI. Data are presented as mean±SD. No significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; CDDP, cisplatin; DCs, dendritic cells; DTX, docetaxel; ELISPOT, enzyme-linked immunospot; FACS, fluorescence-activated cell sorting; GEM, gemcitabine; HMGB-1, high mobility group box-1; IFN-γ, interferon-gamma; LPS, lipopolysaccharide; Luc, luciferase; MHC, major histocompatibility complex; PBS, phosphate-buffered saline; PTX, paclitaxel.

Figure 3

High-dose SQCLC monochemotherapy induce greater…

Figure 3

High-dose SQCLC monochemotherapy induce greater immunosuppression compared with low-dose regimens in vivo. (A)…

Figure 3
High-dose SQCLC monochemotherapy induce greater immunosuppression compared with low-dose regimens in vivo. (A) Chemotherapy treatment scheme. Syngeneic mouse models were established as mentioned above. When tumors became palpable, mice were randomly divided into nine groups receiving an intraperitoneal injection of low-dose CDDP 2.8 mg/kg, high-dose CDDP 8.4 mg/kg, low-dose GEM 60 mg/kg, high-dose GEM 240 mg/kg, low-dose PTX 11 mg/kg, high-dose PTX 33 mg/kg, low-dose DTX 11 mg/kg, high-dose DTX 33 mg/kg, or vehicle. (B) After 30 hours, tumors were harvested for FACS analysis. (C, D) Patients with reatment-naïve SQCLC received either MTD albumin-PTX (at day 0)+nivolumab (routine administration) or low-dose albumin-PTX (1/2 MTD at day 0 and day 7)+nivolumab (routine administration). Before treatment (day 0), baseline PBMCs were evaluated by FACS. At day 7 (before second low-dose albumin-PTX), PBMCs were again assessed by FACS. Cell fractions (cytotoxic T cells and Treg cells) before and after treatment were then compared. Data are presented as mean±SD. N.S., no significance, *p

Figure 4

Low-dose SQCLC monochemotherapy elicits an…

Figure 4

Low-dose SQCLC monochemotherapy elicits an active TIME in vivo. (A) Chemotherapy treatment scheme.…

Figure 4
Low-dose SQCLC monochemotherapy elicits an active TIME in vivo. (A) Chemotherapy treatment scheme. The syngeneic mouse models were established as previously described. When the tumors were palpable, mice were randomly divided into five groups and then received an intraperitoneal injection of CDDP 2.8 mg/kg, GEM 80 mg/kg, DTX 11 mg/kg, PTX 11 mg/kg, or vehicle. (B) After 24 hours, the tumor and spleen tissues were harvested for FACS analysis. Data are presented as mean±SD. N.S., no significance, *p

Figure 5

Combined with anti-PD-1 mAb, sequential…

Figure 5

Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than…

Figure 5
Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than MTD CDDP. (A) Treatment scheme. Upfront low-dose CDDP (2.8 mg/kg, Q7D), MTD CDDP (8.4 mg/kg, Q14D), low-dose or MTD CDDP combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, and vehicle control were injected intraperitoneally (I.P.). (B) Tumor growth curves during treatment. (C, D) Tumor weight and specimens after treatment. (E) Body weight changes during treatment. (F) Tumor growth monitored by BLI. (G–L) Single-cell tumor suspensions were used for FACS to evaluate intratumoral CD45+ CD3+ cells, CD45+ CD3+ CD8+ cells, CD45+ CD3+ CD8+ PD-1+ cells, CD11c+ CD83+ cells, and CD11c+ CD86+ cells. (M–Q) ALT and AST in serum, RBCs, HGB, and PLT in whole blood after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. ALT, alanine transaminase; AST, aspartate aminotransferase; BLI, bioluminescence imaging; CDDP, cisplatin; FACS, fluorescence-activated cell sorting; HGB, hemoglobin; mAb, monoclonal antibody; MTD, maximum tolerated dose; PD-1, programmed death 1; PLT, platelet; RBC, red blood cell.

Figure 6

Combined with anti-PD-1 mAb, upfront…

Figure 6

Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than…

Figure 6
Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than routine MTD DTX. (A) Treatment scheme. Upfront low-dose metronomic DTX (10 mg/kg, Q4D), routine MTD DTX (33 mg/kg, Q7D), equivalent to the standard dose administered to patients, low-dose or MTD DTX combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, or vehicle control were injected intraperitoneally (I.P.) in mice. (B) Body weight changes during treatment. (C) Tumor growth curves during treatment. (D, E) Tumor specimens and weight after treatment. (F) Tumor growth monitored by BLI. (G–L) After treatment, single-cell tumor suspensions were used for FACS. (G) CD11b+ F4/80+ cells (TAMs). (H) CD11b+ F4/80+ MHC-II+cells (type I TAMs). (I) CD11b+ F4/80+ CD206+ cells (type II TAMs). (J) CD11c+ CD83+ cells (activated DCs). (K) CD11c+ CD86+ cells (activated DCs). (L) CD11c+ MHC-II+ cells (activated DCs). (M, N) Mice harboring established OVA-expressing KLN-205 tumors were treated with low-dose+anti-PD-1 mAb, high-dose DTX+anti-PD-1 mAb, or vehicle control by I.P. injection. After 3-week treatment, homolateral tumor-draining lymph nodes were prepared into single-cell suspensions, and subjected to ELISPOT assays. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DCs, dendritic cells; DTX, docetaxel; ELISPOT, enzyme-linked immunospot; FACS, fluorescence-activated cell sorting; Luc, luciferase; mAb, monoclonal antibody; MHC, major histocompatibility complex; MTD, maximum tolerated dose; PD-1, programmed death 1; TAMs, type I tumor-associated macrophages.

Figure 7

Macrophages, CD8 + T cells,…

Figure 7

Macrophages, CD8 + T cells, and gut microbiota may contribute to the synergistic…

Figure 7
Macrophages, CD8+ T cells, and gut microbiota may contribute to the synergistic antitumor effects of DTX plus anti-PD-1 mAb. (A) Schematic of dosing schedule. Macrophage depletion (clodronate, neutral clodronate liposomes, 100 µL per mouse, Q3D, intraperitonially (I.P.)) and CD8+ T cell depletion (in vivo anti-CD8α mAb) treatments were conducted 5 days prior to initiation of the experiment. The treatment groups included DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-1 mAb (10 mg/kg, Q3D, I.P.), DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-L1 mAb (10 mg/kg, Q3D, I.P.), or negative control (vehicle, Q3D, I.P.). (B) Tumor growth monitored by BLI. (C) Tumors dissected from the SQCLC models after treatment. (D) Tumor growth curves during treatment, monitored using vernier calipers. (E) Tumor weight after treatment. (F) Intestinal gut microbiota diversity after treatment. (G) Microbial composition after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DTX, docetaxel; Luc, luciferase; mAb, monoclonal antibody; PD-1, programmed death 1; PD-L1, programmed death-ligand 1; SQCLC, squamous cell lung carcinoma.
All figures (7)
Similar articles
Cited by
References
    1. Bertaglia V, Vallone S, Pacchiana MV, et al. . Advanced squamous lung cancer: therapeutic options, future directions, unmet needs and results of a monocentric survey. Lung Cancer Manag 2017;6:93–107. 10.2217/lmt-2017-0011 - DOI - PMC - PubMed
    1. Bunn PA. Karnofsky Award 2016: a lung cancer journey, 1973 to 2016. J Clin Oncol 2017;35:243–52. 10.1200/JCO.2016.70.4064 - DOI - PubMed
    1. Schiller JH, Harrington D, Belani CP, et al. . Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92–8. 10.1056/NEJMoa011954 - DOI - PubMed
    1. Cheng H, Shcherba M, Kandavelou K, et al. . Emerging drugs for squamous cell lung cancer. Expert Opin Emerg Drugs 2015;20:149–60. 10.1517/14728214.2015.1001365 - DOI - PubMed
    1. Lazzari C, Karachaliou N, Gregorc V, et al. . Second-Line therapy of squamous non-small cell lung cancer: an evolving landscape. Expert Rev Respir Med 2017;11:469–79. 10.1080/17476348.2017.1326822 - DOI - PubMed
Show all 44 references
Publication types
MeSH terms
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2
Figure 2
Low-dose chemotherapy induces HMGB-1 release by SQCLC cells, thereby promoting DC maturation (A–D) and inducing systemic immune responses as well as immunological memory in vivo (E, F). KLN 205-Luc cells were incubated with 4 µM CDDP, 20 µM GEM, 1 µM DTX, or 1 µM PTX for 30 hours, after which the conditioned media were dialyzed at 4°C for 24 hours using a 10,000-MWCO dialysis tubing to remove any residual chemotherapeutic drug. Immature murine bone marrow-derived dendritic cells (BMDCs) cultured in 6-well plates (1×106 cells/mL) were then cultured in unconditioned media (negative control), media containing LPS (10 µg/mL, positive control), or different conditioned media, and were harvested 24 hours later. The maturation state of BMDCs was evaluated by measuring the cell surface co-expression of CD11c+ CD11b+ CD80+, CD11c+ CD11b+ CD86+, and CD11c+ CD11b+ MHC class II+ by FACS. (A) percentage of BMDCs expressing the above-described combinations of three different cell maturation biomarkers after the different stimuli, as revealed by FACS. (B) Proportion of total mature BMDCs in each group after the different stimuli. (C) HMGB-1 levels in the media after treatment with different chemotherapeutic agents for 30 hours, as assessed by ELISA. (D) Immature BMDCs were stimulated with 50 ng/mL or 100 ng/mL recombinant HMGB-1 for 24 hours, and FACS analysis of CD11c+ CD11b+ CD80+, CD11c+ CD11b+ CD86+, CD11c+ CD11b+ MHC class II+ expression was carried out. (E, F) Murine KLN-205-Luc cells were pretreated with 1 µM CDDP, 15 µM CDDP for 72 hours or with PBS as the negative control. These pretreated cells (1×106) were subcutaneously injected into the right flanks of mice. Splenocytes were harvested after 7 days, and IFN-γ secretion was evaluated using ELISPOT, according to the kit manufacturer’s protocol. (G) The aforementioned mice were rechallenged with 2×105 live KLN-205 cells in the opposite flank, and tumor growth was monitored using vernier calipers and BLI. Data are presented as mean±SD. No significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; CDDP, cisplatin; DCs, dendritic cells; DTX, docetaxel; ELISPOT, enzyme-linked immunospot; FACS, fluorescence-activated cell sorting; GEM, gemcitabine; HMGB-1, high mobility group box-1; IFN-γ, interferon-gamma; LPS, lipopolysaccharide; Luc, luciferase; MHC, major histocompatibility complex; PBS, phosphate-buffered saline; PTX, paclitaxel.
Figure 3
Figure 3
High-dose SQCLC monochemotherapy induce greater immunosuppression compared with low-dose regimens in vivo. (A) Chemotherapy treatment scheme. Syngeneic mouse models were established as mentioned above. When tumors became palpable, mice were randomly divided into nine groups receiving an intraperitoneal injection of low-dose CDDP 2.8 mg/kg, high-dose CDDP 8.4 mg/kg, low-dose GEM 60 mg/kg, high-dose GEM 240 mg/kg, low-dose PTX 11 mg/kg, high-dose PTX 33 mg/kg, low-dose DTX 11 mg/kg, high-dose DTX 33 mg/kg, or vehicle. (B) After 30 hours, tumors were harvested for FACS analysis. (C, D) Patients with reatment-naïve SQCLC received either MTD albumin-PTX (at day 0)+nivolumab (routine administration) or low-dose albumin-PTX (1/2 MTD at day 0 and day 7)+nivolumab (routine administration). Before treatment (day 0), baseline PBMCs were evaluated by FACS. At day 7 (before second low-dose albumin-PTX), PBMCs were again assessed by FACS. Cell fractions (cytotoxic T cells and Treg cells) before and after treatment were then compared. Data are presented as mean±SD. N.S., no significance, *p

Figure 4

Low-dose SQCLC monochemotherapy elicits an…

Figure 4

Low-dose SQCLC monochemotherapy elicits an active TIME in vivo. (A) Chemotherapy treatment scheme.…

Figure 4
Low-dose SQCLC monochemotherapy elicits an active TIME in vivo. (A) Chemotherapy treatment scheme. The syngeneic mouse models were established as previously described. When the tumors were palpable, mice were randomly divided into five groups and then received an intraperitoneal injection of CDDP 2.8 mg/kg, GEM 80 mg/kg, DTX 11 mg/kg, PTX 11 mg/kg, or vehicle. (B) After 24 hours, the tumor and spleen tissues were harvested for FACS analysis. Data are presented as mean±SD. N.S., no significance, *p

Figure 5

Combined with anti-PD-1 mAb, sequential…

Figure 5

Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than…

Figure 5
Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than MTD CDDP. (A) Treatment scheme. Upfront low-dose CDDP (2.8 mg/kg, Q7D), MTD CDDP (8.4 mg/kg, Q14D), low-dose or MTD CDDP combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, and vehicle control were injected intraperitoneally (I.P.). (B) Tumor growth curves during treatment. (C, D) Tumor weight and specimens after treatment. (E) Body weight changes during treatment. (F) Tumor growth monitored by BLI. (G–L) Single-cell tumor suspensions were used for FACS to evaluate intratumoral CD45+ CD3+ cells, CD45+ CD3+ CD8+ cells, CD45+ CD3+ CD8+ PD-1+ cells, CD11c+ CD83+ cells, and CD11c+ CD86+ cells. (M–Q) ALT and AST in serum, RBCs, HGB, and PLT in whole blood after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. ALT, alanine transaminase; AST, aspartate aminotransferase; BLI, bioluminescence imaging; CDDP, cisplatin; FACS, fluorescence-activated cell sorting; HGB, hemoglobin; mAb, monoclonal antibody; MTD, maximum tolerated dose; PD-1, programmed death 1; PLT, platelet; RBC, red blood cell.

Figure 6

Combined with anti-PD-1 mAb, upfront…

Figure 6

Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than…

Figure 6
Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than routine MTD DTX. (A) Treatment scheme. Upfront low-dose metronomic DTX (10 mg/kg, Q4D), routine MTD DTX (33 mg/kg, Q7D), equivalent to the standard dose administered to patients, low-dose or MTD DTX combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, or vehicle control were injected intraperitoneally (I.P.) in mice. (B) Body weight changes during treatment. (C) Tumor growth curves during treatment. (D, E) Tumor specimens and weight after treatment. (F) Tumor growth monitored by BLI. (G–L) After treatment, single-cell tumor suspensions were used for FACS. (G) CD11b+ F4/80+ cells (TAMs). (H) CD11b+ F4/80+ MHC-II+cells (type I TAMs). (I) CD11b+ F4/80+ CD206+ cells (type II TAMs). (J) CD11c+ CD83+ cells (activated DCs). (K) CD11c+ CD86+ cells (activated DCs). (L) CD11c+ MHC-II+ cells (activated DCs). (M, N) Mice harboring established OVA-expressing KLN-205 tumors were treated with low-dose+anti-PD-1 mAb, high-dose DTX+anti-PD-1 mAb, or vehicle control by I.P. injection. After 3-week treatment, homolateral tumor-draining lymph nodes were prepared into single-cell suspensions, and subjected to ELISPOT assays. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DCs, dendritic cells; DTX, docetaxel; ELISPOT, enzyme-linked immunospot; FACS, fluorescence-activated cell sorting; Luc, luciferase; mAb, monoclonal antibody; MHC, major histocompatibility complex; MTD, maximum tolerated dose; PD-1, programmed death 1; TAMs, type I tumor-associated macrophages.

Figure 7

Macrophages, CD8 + T cells,…

Figure 7

Macrophages, CD8 + T cells, and gut microbiota may contribute to the synergistic…

Figure 7
Macrophages, CD8+ T cells, and gut microbiota may contribute to the synergistic antitumor effects of DTX plus anti-PD-1 mAb. (A) Schematic of dosing schedule. Macrophage depletion (clodronate, neutral clodronate liposomes, 100 µL per mouse, Q3D, intraperitonially (I.P.)) and CD8+ T cell depletion (in vivo anti-CD8α mAb) treatments were conducted 5 days prior to initiation of the experiment. The treatment groups included DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-1 mAb (10 mg/kg, Q3D, I.P.), DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-L1 mAb (10 mg/kg, Q3D, I.P.), or negative control (vehicle, Q3D, I.P.). (B) Tumor growth monitored by BLI. (C) Tumors dissected from the SQCLC models after treatment. (D) Tumor growth curves during treatment, monitored using vernier calipers. (E) Tumor weight after treatment. (F) Intestinal gut microbiota diversity after treatment. (G) Microbial composition after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DTX, docetaxel; Luc, luciferase; mAb, monoclonal antibody; PD-1, programmed death 1; PD-L1, programmed death-ligand 1; SQCLC, squamous cell lung carcinoma.
All figures (7)
Similar articles
Cited by
References
    1. Bertaglia V, Vallone S, Pacchiana MV, et al. . Advanced squamous lung cancer: therapeutic options, future directions, unmet needs and results of a monocentric survey. Lung Cancer Manag 2017;6:93–107. 10.2217/lmt-2017-0011 - DOI - PMC - PubMed
    1. Bunn PA. Karnofsky Award 2016: a lung cancer journey, 1973 to 2016. J Clin Oncol 2017;35:243–52. 10.1200/JCO.2016.70.4064 - DOI - PubMed
    1. Schiller JH, Harrington D, Belani CP, et al. . Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92–8. 10.1056/NEJMoa011954 - DOI - PubMed
    1. Cheng H, Shcherba M, Kandavelou K, et al. . Emerging drugs for squamous cell lung cancer. Expert Opin Emerg Drugs 2015;20:149–60. 10.1517/14728214.2015.1001365 - DOI - PubMed
    1. Lazzari C, Karachaliou N, Gregorc V, et al. . Second-Line therapy of squamous non-small cell lung cancer: an evolving landscape. Expert Rev Respir Med 2017;11:469–79. 10.1080/17476348.2017.1326822 - DOI - PubMed
Show all 44 references
Publication types
MeSH terms
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Low-dose SQCLC monochemotherapy elicits an active TIME in vivo. (A) Chemotherapy treatment scheme. The syngeneic mouse models were established as previously described. When the tumors were palpable, mice were randomly divided into five groups and then received an intraperitoneal injection of CDDP 2.8 mg/kg, GEM 80 mg/kg, DTX 11 mg/kg, PTX 11 mg/kg, or vehicle. (B) After 24 hours, the tumor and spleen tissues were harvested for FACS analysis. Data are presented as mean±SD. N.S., no significance, *p

Figure 5

Combined with anti-PD-1 mAb, sequential…

Figure 5

Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than…

Figure 5
Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than MTD CDDP. (A) Treatment scheme. Upfront low-dose CDDP (2.8 mg/kg, Q7D), MTD CDDP (8.4 mg/kg, Q14D), low-dose or MTD CDDP combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, and vehicle control were injected intraperitoneally (I.P.). (B) Tumor growth curves during treatment. (C, D) Tumor weight and specimens after treatment. (E) Body weight changes during treatment. (F) Tumor growth monitored by BLI. (G–L) Single-cell tumor suspensions were used for FACS to evaluate intratumoral CD45+ CD3+ cells, CD45+ CD3+ CD8+ cells, CD45+ CD3+ CD8+ PD-1+ cells, CD11c+ CD83+ cells, and CD11c+ CD86+ cells. (M–Q) ALT and AST in serum, RBCs, HGB, and PLT in whole blood after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. ALT, alanine transaminase; AST, aspartate aminotransferase; BLI, bioluminescence imaging; CDDP, cisplatin; FACS, fluorescence-activated cell sorting; HGB, hemoglobin; mAb, monoclonal antibody; MTD, maximum tolerated dose; PD-1, programmed death 1; PLT, platelet; RBC, red blood cell.

Figure 6

Combined with anti-PD-1 mAb, upfront…

Figure 6

Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than…

Figure 6
Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than routine MTD DTX. (A) Treatment scheme. Upfront low-dose metronomic DTX (10 mg/kg, Q4D), routine MTD DTX (33 mg/kg, Q7D), equivalent to the standard dose administered to patients, low-dose or MTD DTX combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, or vehicle control were injected intraperitoneally (I.P.) in mice. (B) Body weight changes during treatment. (C) Tumor growth curves during treatment. (D, E) Tumor specimens and weight after treatment. (F) Tumor growth monitored by BLI. (G–L) After treatment, single-cell tumor suspensions were used for FACS. (G) CD11b+ F4/80+ cells (TAMs). (H) CD11b+ F4/80+ MHC-II+cells (type I TAMs). (I) CD11b+ F4/80+ CD206+ cells (type II TAMs). (J) CD11c+ CD83+ cells (activated DCs). (K) CD11c+ CD86+ cells (activated DCs). (L) CD11c+ MHC-II+ cells (activated DCs). (M, N) Mice harboring established OVA-expressing KLN-205 tumors were treated with low-dose+anti-PD-1 mAb, high-dose DTX+anti-PD-1 mAb, or vehicle control by I.P. injection. After 3-week treatment, homolateral tumor-draining lymph nodes were prepared into single-cell suspensions, and subjected to ELISPOT assays. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DCs, dendritic cells; DTX, docetaxel; ELISPOT, enzyme-linked immunospot; FACS, fluorescence-activated cell sorting; Luc, luciferase; mAb, monoclonal antibody; MHC, major histocompatibility complex; MTD, maximum tolerated dose; PD-1, programmed death 1; TAMs, type I tumor-associated macrophages.

Figure 7

Macrophages, CD8 + T cells,…

Figure 7

Macrophages, CD8 + T cells, and gut microbiota may contribute to the synergistic…

Figure 7
Macrophages, CD8+ T cells, and gut microbiota may contribute to the synergistic antitumor effects of DTX plus anti-PD-1 mAb. (A) Schematic of dosing schedule. Macrophage depletion (clodronate, neutral clodronate liposomes, 100 µL per mouse, Q3D, intraperitonially (I.P.)) and CD8+ T cell depletion (in vivo anti-CD8α mAb) treatments were conducted 5 days prior to initiation of the experiment. The treatment groups included DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-1 mAb (10 mg/kg, Q3D, I.P.), DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-L1 mAb (10 mg/kg, Q3D, I.P.), or negative control (vehicle, Q3D, I.P.). (B) Tumor growth monitored by BLI. (C) Tumors dissected from the SQCLC models after treatment. (D) Tumor growth curves during treatment, monitored using vernier calipers. (E) Tumor weight after treatment. (F) Intestinal gut microbiota diversity after treatment. (G) Microbial composition after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DTX, docetaxel; Luc, luciferase; mAb, monoclonal antibody; PD-1, programmed death 1; PD-L1, programmed death-ligand 1; SQCLC, squamous cell lung carcinoma.
All figures (7)
Figure 5
Figure 5
Combined with anti-PD-1 mAb, sequential low-dose CDDP exerts greater synergistic antitumor effects than MTD CDDP. (A) Treatment scheme. Upfront low-dose CDDP (2.8 mg/kg, Q7D), MTD CDDP (8.4 mg/kg, Q14D), low-dose or MTD CDDP combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, and vehicle control were injected intraperitoneally (I.P.). (B) Tumor growth curves during treatment. (C, D) Tumor weight and specimens after treatment. (E) Body weight changes during treatment. (F) Tumor growth monitored by BLI. (G–L) Single-cell tumor suspensions were used for FACS to evaluate intratumoral CD45+ CD3+ cells, CD45+ CD3+ CD8+ cells, CD45+ CD3+ CD8+ PD-1+ cells, CD11c+ CD83+ cells, and CD11c+ CD86+ cells. (M–Q) ALT and AST in serum, RBCs, HGB, and PLT in whole blood after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. ALT, alanine transaminase; AST, aspartate aminotransferase; BLI, bioluminescence imaging; CDDP, cisplatin; FACS, fluorescence-activated cell sorting; HGB, hemoglobin; mAb, monoclonal antibody; MTD, maximum tolerated dose; PD-1, programmed death 1; PLT, platelet; RBC, red blood cell.
Figure 6
Figure 6
Combined with anti-PD-1 mAb, upfront metronomic DTX exerts greater tumor inhibitory effects than routine MTD DTX. (A) Treatment scheme. Upfront low-dose metronomic DTX (10 mg/kg, Q4D), routine MTD DTX (33 mg/kg, Q7D), equivalent to the standard dose administered to patients, low-dose or MTD DTX combined with an anti-PD-1 mAb, anti-PD-1 mAb alone, or vehicle control were injected intraperitoneally (I.P.) in mice. (B) Body weight changes during treatment. (C) Tumor growth curves during treatment. (D, E) Tumor specimens and weight after treatment. (F) Tumor growth monitored by BLI. (G–L) After treatment, single-cell tumor suspensions were used for FACS. (G) CD11b+ F4/80+ cells (TAMs). (H) CD11b+ F4/80+ MHC-II+cells (type I TAMs). (I) CD11b+ F4/80+ CD206+ cells (type II TAMs). (J) CD11c+ CD83+ cells (activated DCs). (K) CD11c+ CD86+ cells (activated DCs). (L) CD11c+ MHC-II+ cells (activated DCs). (M, N) Mice harboring established OVA-expressing KLN-205 tumors were treated with low-dose+anti-PD-1 mAb, high-dose DTX+anti-PD-1 mAb, or vehicle control by I.P. injection. After 3-week treatment, homolateral tumor-draining lymph nodes were prepared into single-cell suspensions, and subjected to ELISPOT assays. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DCs, dendritic cells; DTX, docetaxel; ELISPOT, enzyme-linked immunospot; FACS, fluorescence-activated cell sorting; Luc, luciferase; mAb, monoclonal antibody; MHC, major histocompatibility complex; MTD, maximum tolerated dose; PD-1, programmed death 1; TAMs, type I tumor-associated macrophages.
Figure 7
Figure 7
Macrophages, CD8+ T cells, and gut microbiota may contribute to the synergistic antitumor effects of DTX plus anti-PD-1 mAb. (A) Schematic of dosing schedule. Macrophage depletion (clodronate, neutral clodronate liposomes, 100 µL per mouse, Q3D, intraperitonially (I.P.)) and CD8+ T cell depletion (in vivo anti-CD8α mAb) treatments were conducted 5 days prior to initiation of the experiment. The treatment groups included DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-1 mAb (10 mg/kg, Q3D, I.P.), DTX (10 mg/kg, Q3D, I.P.) combined with anti-PD-L1 mAb (10 mg/kg, Q3D, I.P.), or negative control (vehicle, Q3D, I.P.). (B) Tumor growth monitored by BLI. (C) Tumors dissected from the SQCLC models after treatment. (D) Tumor growth curves during treatment, monitored using vernier calipers. (E) Tumor weight after treatment. (F) Intestinal gut microbiota diversity after treatment. (G) Microbial composition after treatment. Data are presented as mean±SD. N.S., no significance, *p<0.05; **p<0.01; ***p<0.001. BLI, bioluminescence imaging; DTX, docetaxel; Luc, luciferase; mAb, monoclonal antibody; PD-1, programmed death 1; PD-L1, programmed death-ligand 1; SQCLC, squamous cell lung carcinoma.

References

    1. Bertaglia V, Vallone S, Pacchiana MV, et al. . Advanced squamous lung cancer: therapeutic options, future directions, unmet needs and results of a monocentric survey. Lung Cancer Manag 2017;6:93–107. 10.2217/lmt-2017-0011
    1. Bunn PA. Karnofsky Award 2016: a lung cancer journey, 1973 to 2016. J Clin Oncol 2017;35:243–52. 10.1200/JCO.2016.70.4064
    1. Schiller JH, Harrington D, Belani CP, et al. . Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92–8. 10.1056/NEJMoa011954
    1. Cheng H, Shcherba M, Kandavelou K, et al. . Emerging drugs for squamous cell lung cancer. Expert Opin Emerg Drugs 2015;20:149–60. 10.1517/14728214.2015.1001365
    1. Lazzari C, Karachaliou N, Gregorc V, et al. . Second-Line therapy of squamous non-small cell lung cancer: an evolving landscape. Expert Rev Respir Med 2017;11:469–79. 10.1080/17476348.2017.1326822
    1. Fritz JM, Lenardo MJ. Development of immune checkpoint therapy for cancer. J Exp Med 2019;216:1244–54. 10.1084/jem.20182395
    1. Heinhuis KM, Ros W, Kok M, et al. . Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors. Ann Oncol 2019;30:219–35. 10.1093/annonc/mdy551
    1. Wilky BA. Immune checkpoint inhibitors: the linchpins of modern immunotherapy. Immunol Rev 2019;290:6–23. 10.1111/imr.12766
    1. Paz-Ares L, Luft A, Vicente D, et al. . Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med 2018;379:2040–51. 10.1056/NEJMoa1810865
    1. Jotte RM, Cappuzzo F, Vynnychenko I, et al. . IMpower131: primary pfs and safety analysis of a randomized phase III study of atezolizumab + carboplatin + paclitaxel or nab-paclitaxel vs carboplatin + nab-paclitaxel as 1L therapy in advanced squamous NSCLC. JCO 2018;36:LBA9000 10.1200/JCO.2018.36.18_suppl.LBA9000
    1. Peters S, Reck M, Smit EF, et al. . How to make the best use of immunotherapy as first-line treatment of advanced/metastatic non-small-cell lung cancer. Ann Oncol 2019;30:884–96. 10.1093/annonc/mdz109
    1. Mathios D, Kim JE, Mangraviti A, et al. . Anti-Pd-1 antitumor immunity is enhanced by local and abrogated by systemic chemotherapy in GBM. Sci Transl Med 2016;8:370ra180. 10.1126/scitranslmed.aag2942
    1. Galluzzi L, Buqué A, Kepp O, et al. . Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 2015;28:690–714. 10.1016/j.ccell.2015.10.012
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA A Cancer J Clin 2019;69:7–34. 10.3322/caac.21551
    1. Shafique M, Tanvetyanon T. Immunotherapy alone or chemo-immunotherapy as front-line treatment for advanced non-small cell lung cancer. Expert Opin Biol Ther 2019;19:225–32. 10.1080/14712598.2019.1571036
    1. Nosaki K, Saka H, Hosomi Y, et al. . Safety and efficacy of pembrolizumab monotherapy in elderly patients with PD-L1-positive advanced non-small-cell lung cancer: pooled analysis from the KEYNOTE-010, KEYNOTE-024, and KEYNOTE-042 studies. Lung Cancer 2019;135:188–95. 10.1016/j.lungcan.2019.07.004
    1. Okita R, Yukawa T, Nojima Y, et al. . Mhc class I chain-related molecule A and B expression is upregulated by cisplatin and associated with good prognosis in patients with non-small cell lung cancer. Cancer Immunol Immunother 2016;65:499–509. 10.1007/s00262-016-1814-9
    1. Zheng H, Zeltsman M, Zauderer MG, et al. . Chemotherapy-Induced immunomodulation in non-small-cell lung cancer: a rationale for combination chemoimmunotherapy. Immunotherapy 2017;9:913–27. 10.2217/imt-2017-0052
    1. de Biasi AR, Villena-Vargas J, Adusumilli PS. Cisplatin-Induced antitumor immunomodulation: a review of preclinical and clinical evidence. Clin Cancer Res 2014;20:5384–91. 10.1158/1078-0432.CCR-14-1298
    1. Cullis J, Siolas D, Avanzi A, et al. . Macropinocytosis of nab-paclitaxel drives macrophage activation in pancreatic cancer. Cancer Immunol Res 2017;5:182–90. 10.1158/2326-6066.CIR-16-0125
    1. Liu WM, Fowler DW, Smith P, et al. . Pre-Treatment with chemotherapy can enhance the antigenicity and immunogenicity of tumours by promoting adaptive immune responses. Br J Cancer 2010;102:115–23. 10.1038/sj.bjc.6605465
    1. Galluzzi L, Humeau J, Buqué A, et al. . Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol 2020:bbaa184. 10.1038/s41571-020-0413-z
    1. Pfirschke C, Engblom C, Rickelt S, et al. . Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity 2016;44:343–54. 10.1016/j.immuni.2015.11.024
    1. Song W, Shen L, Wang Y, et al. . Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap. Nat Commun 2018;9:2237. 10.1038/s41467-018-04605-x
    1. Weir GM, Hrytsenko O, Stanford MM, et al. . Metronomic cyclophosphamide enhances HPV16E7 peptide vaccine induced antigen-specific and cytotoxic T-cell mediated antitumor immune response. Oncoimmunology 2014;3:e953407. 10.4161/21624011.2014.953407
    1. Denies S, Cicchelero L, Van Audenhove I, et al. . Combination of interleukin-12 gene therapy, metronomic cyclophosphamide and DNA cancer vaccination directs all arms of the immune system towards tumor eradication. J Control Release 2014;187:175–82. 10.1016/j.jconrel.2014.05.045
    1. Liu X, Pu Y, Cron K, et al. . Cd47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat Med 2015;21:1209–15. 10.1038/nm.3931
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197–218. 10.1038/s41573-018-0007-y
    1. Serrano-Del Valle A, Anel A, Naval J, et al. . Immunogenic cell death and immunotherapy of multiple myeloma. Front Cell Dev Biol 2019;7:50. 10.3389/fcell.2019.00050
    1. Caamaño J, Hunter CA. Nf-kappaB family of transcription factors: central regulators of innate and adaptive immune functions. Clin Microbiol Rev 2002;15:414–29. 10.1128/CMR.15.3.414-429.2002
    1. Jin X, Ding D, Yan Y, et al. . Phosphorylated Rb promotes cancer immunity by inhibiting NF-κB activation and PD-L1 expression. Mol Cell 2019;73:22–35. e26. 10.1016/j.molcel.2018.10.034
    1. Ramakrishnan SK, Zhang H, Ma X, et al. . Intestinal non-canonical NFκB signaling shapes the local and systemic immune response. Nat Commun 2019;10:660. 10.1038/s41467-019-08581-8
    1. Constantino J, Gomes C, Falcão A, et al. . Dendritic cell-based immunotherapy: a basic review and recent advances. Immunol Res 2017;65:798–810. 10.1007/s12026-017-8931-1
    1. Pandolfi F, Altamura S, Frosali S, et al. . Key role of dAMP in inflammation, cancer, and tissue repair. Clin Ther 2016;38:1017–28. 10.1016/j.clinthera.2016.02.028
    1. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. Faseb J 2008;22:659–61. 10.1096/fj.07-9574LSF
    1. Laksono BM, de Vries RD, Verburgh RJ, et al. . Studies into the mechanism of Measles-associated immune suppression during a measles outbreak in the Netherlands. Nat Commun 2018;9:4944. 10.1038/s41467-018-07515-0
    1. Gridelli C, Gallo C, Di Maio M, et al. . A randomised clinical trial of two docetaxel regimens (Weekly vs 3 week) in the second-line treatment of non-small-cell lung cancer. the distal 01 study. Br J Cancer 2004;91:1996–2004. 10.1038/sj.bjc.6602241
    1. Di Maio M, Perrone F, Chiodini P, et al. . Individual patient data meta-analysis of docetaxel administered once every 3 weeks compared with once every week second-line treatment of advanced non-small-cell lung cancer. J Clin Oncol 2007;25:1377–82. 10.1200/JCO.2006.09.8251
    1. Voorwerk L, Slagter M, Horlings HM, et al. . Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the tonic trial. Nat Med 2019;25:920–8. 10.1038/s41591-019-0432-4
    1. Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer 2017;17:271–85. 10.1038/nrc.2017.13
    1. Dzutsev A, Goldszmid RS, Viaud S, et al. . The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol 2015;45:17–31. 10.1002/eji.201444972
    1. Iida N, Dzutsev A, Stewart CA, et al. . Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013;342:967–70. 10.1126/science.1240527
    1. Vétizou M, Pitt JM, Daillère R, et al. . Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015;350:1079–84. 10.1126/science.aad1329
    1. Sivan A, Corrales L, Hubert N, et al. . Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015;350:1084–9. 10.1126/science.aac4255

Source: PubMed

3
Předplatit