Biological and Pharmaceutical Bulletin
Online ISSN : 1347-5215
Print ISSN : 0918-6158
ISSN-L : 0918-6158
Regular Articles
Increasing Tumor Extracellular pH by an Oral Alkalinizing Agent Improves Antitumor Responses of Anti-PD-1 Antibody: Implication of Relationships between Serum Bicarbonate Concentrations, Urinary pH, and Therapeutic Outcomes
Hidenori AndoSherif E. EmamYoshino KawaguchiTaro ShimizuYu IshimaKiyoshi EshimaTatsuhiro Ishida
Author information
JOURNAL FREE ACCESS FULL-TEXT HTML

2021 Volume 44 Issue 6 Pages 844-852

Details
Abstract

Acidic extracellular pH (pHe) is characteristic of the tumor microenvironment. Several reports suggest that increasing pHe improves the response of immune checkpoint inhibitors in murine models. To increase pHe, either sodium bicarbonate (NaHCO3) or citric acid/potassium-sodium citrate (KNa-cit) was chronically administered to mice. It is hypothesized that bicarbonate ions (HCO3), produced from these alkalinizing agents in vivo, increased pHe in the tumor, and excess HCO3 eliminated into urine increased urinary pH values. However, there is little published information on the effect of changing serum HCO3 concentrations, urinary HCO3 concentrations and urinary pH values on the therapeutic outcomes of immunotherapy. In this study, we report that oral administration of either NaHCO3 or KNa-cit increased responses to anti-programmed cell death-1 (PD-1) antibody, an immune checkpoint inhibitor, in a murine B16 melanoma model. In addition, we report that daily oral administration of an alkalinizing agent increased blood HCO3 concentrations, corresponding to increasing the tumor pHe. Serum HCO3 concentrations also correlated with urinary HCO3 concentrations and urinary pH values. There was a clear relationship between urinary pH values and the antitumor effects of immunotherapy with anti-PD-1 antibody. Our results imply that blood HCO3 concentrations, corresponding to tumor pHe and urinary pH values, may be important factors that predict the clinical outcomes of an immunotherapeutic agent, when combined with alkalinizing agents such as NaHCO3 and KNa-cit.

INTRODUCTION

Several reports have demonstrated that the interstitial extracellular pH (pHe) of tumors is acidic (pH 6.2–6.9) compared to normal tissues (pH 7.3–7.4) and the intracellular pH of tumor cells (pHi) can be more alkaline (pH 7.1–7.7) than normal cells (pH 7.0–7.2),13) which creates a reversed extracellular to intracellular pH gradient in tumors relative to normal tissues. This reversed pH gradient is produced partly by the overstimulation of several ion transporters such as the Na+/H+ exchanger (NHE-1), the Na+-independent and Na+-dependent HCO3/Cl exchangers, and the H+/lactate cotransporter (the monocarboxylate transporter; MCT), which secret H+ ions into the extracellular space.4) In addition, tumor-associated anabolic glycolysis, called the Warburg effect, is one of the principal factors leading to the acidic tumor extracellular environment.57) In tumors, the glucose transporter GLUT1, which generates ATP by glycolysis, is overexpressed and several metabolic intermediates such as lactate and pyruvate are generated. Lactate, together with H+ ions, via the action of MCT, is transported to the extracellular fluid, leading to its acidification.8) Many studies reported that the reversed pH gradient across the cellular membrane is strongly related to uncontrolled progression, angiogenesis, and metastasis of tumors.913)

It has been reported that tumor acidity can influence responses to immunotherapy.1416) Acidification of the tumor interstitial space may promote escape of solid tumors from immune surveillance and potentially limit the efficacy of immunotherapies such as an immune checkpoint inhibitor.17,18) Pilon-Thomas et al. reported that increasing tumor pHe with sodium bicarbonate (NaHCO3), an alkalinizing agent, increased responses to immune checkpoint inhibitors in a B16 melanoma murine model, where it was associated with increased T-cell infiltration.17) These proof-of-concept studies suggest that the acidic pHe within the tumor microenvironment is a potential target to improve the response to immunotherapies.

Oral NaHCO3 administration was used in several studies to modulate the acidic pHe in solid tumors.1820) Raghunand et al. have shown that NaHCO3 effectively reversed pH gradients in tumors, but not in normal tissues.21) The in vivo bicarbonate buffer system involves the balance of carbonic acid (H2CO3), HCO3 and carbon dioxide (CO2). It plays an important role in maintaining the homeostatic control of blood pH, and as a consequence, contributes to tumor pHe.22) Oral administration of NaHCO3 would, in theory but never elucidated, increase serum HCO3 concentrations and deliver excess HCO3 into solid tumors. The HCO3 traps a H+ ion in the tumor interstitial space and forms H2CO3, resulting in the increased tumor pHe. The hydration rate of CO2 is much higher than the dehydration rate of H2CO3,23) suggesting that H2CO3 quickly dissociates into CO2 and H2O. Excess CO2 is lost through the lungs. Renal glomerular filtration regulates blood levels of HCO3 and acid secretion.24) We can hypothesize that marked increases of urinary HCO3 concentration and urinary pH values resulting from increases in serum HCO3 will lead to increases in the tumor pHe. To date, however, few reports have examined the effects of an alkalinizing agent on increasing serum HCO3 concentrations, urinary HCO3 concentrations, and urinary pH values, and showed the relationships of these on the therapeutic outcomes of immunotherapies.

In this study, we show that the change in serum HCO3 concentrations and subsequent changes in urinary HCO3 concentrations, urinary pH values, and corresponding tumor pHe, by oral administration of NaHCO3 or citric acid/potassium-sodium citrate (KNa-cit). Oral KNa-cit is a drug used to treat metabolic acidosis,25) and may also be a good means of generating HCO3 and alkalinizing the acidic pHe in solid tumors, because KNa-cit is effectively converted to HCO3 in a body after intestinal absorption.26) In addition, we show the neutralization of tumor pHe by oral administration of alkalinizing agents increases responses to anti-programmed cell death-1 (PD-1) antibodies, an immune checkpoint inhibitor, and the relationship between the change in urinary pH values and the anticancer effects.

MATERIALS AND METHODS

Materials

NaHCO3 and magnesium oxide (MgO) were purchased from KENEI Pharmaceutical (Osaka, Japan). Anti-PD-1 antibody (InVivomAb anti-mouse PD-1 (CD279), clone: RMP1-14) was purchased from Bio X Cell (NH, U.S.A.). All other reagents were of analytical grade.

Cells and Animals

The B16 murine melanoma cells (RCB2638) and Colon-26 murine colorectal carcinoma cells (RCB2657) were purchased from the RIKEN BioResource Center (Ibaraki, Japan). The cells were cultured in RPMI-1640 medium (FUJIFILM Wako Pure Chemical Corp., Osaka, Japan) supplemented with 10% heat-inactivated fetal bovine serum (Corning, NY, U.S.A.), 100 units/mL penicillin and 100 µg/mL streptomycin (ICN Biomedicals, CA, U.S.A.) in a 5% CO2/air incubator at 37 °C.

BALB/c mice (female, 5 weeks old) and C57BL/6 mice (female, 5 weeks old) were purchased from Japan SLC (Shizuoka, Japan). The experimental animals were allowed free access to water and mouse chow, and were housed under controlled environmental conditions (constant temperature and humidity, and a 12-h dark–light cycle). All animal experiments were evaluated and approved by the Animal and Ethics Review Committee of Tokushima University.

Tumor-Bearing Mouse Model

B16 tumor-bearing mouse model was established by subcutaneous inoculation of B16 cells (2 × 106 cells/mouse) in the flank of C57BL/6 mice. Colon-26 tumor-bearing mouse model was established by subcutaneous inoculation of Colon-26 cells (2 × 106 cells/mouse) at a flank region of BALB/c mice. All animal experiments were initiated when the tumors reached about 100 mm3 in size.

Measurement of Serum HCO3, Urinary HCO3 and Urinary pH Values

BALB/c mice were orally administered with a dose of NaHCO3 (500 mg/kg) plus MgO (165 mg/kg) or KNa-cit (500 mg/kg), a citrate mixture composed of 2 mol potassium citrate, 2 mol sodium citrate and 1 mol citric acid. Each reagent was dissolved in water for oral administration. At selected time points post administration (0, 0.5, 1, 2, 4, 6, 8, and 24 h), blood was collected from the postcaval vein of the mice. Serum samples were obtained by centrifugation of the blood (3000 rpm, 4 °C, 15 min) following to incubation for 30 min at room temperature. The serum HCO3 concentrations were measured using the HCO3 measurement kit (Diacolor® CO2 clinical diagnostic reagent, TOYOBO, Osaka, Japan). At the same time points, urine was collected by pushing on the lower abdominal region of the mice. The urinary HCO3 concentrations were measured using the HCO3 measurement kit (Diacolor® CO2). Urinary pH values were measured with the pH test paper (pH 5.5–9.0, AS ONE, Osaka, Japan).

Measurement of Intratumor pH Using a Microelectrode

Colon-26 tumor-bearing mice were allowed to drink water containing 200 mM NaHCO3 ad lib. After 14-d ingestion of NaHCO3 solution, the interstitial pHe in the tumor was measured using microelectrode with pH meter as referred the previous publications.20,27) The mice were anesthetized by isoflurane inhalation and then stabbed with a reference electrode (MI-401F, Microelectrodes, NH, U.S.A.) into the subcutaneous region and with a pH electrode (MI-408B TIP, Microelectrodes) into the center of the tumor. Electrodes were calibrated before the measurements using standard pH 4.01 and 7.00 buffers. One measurement was taken at each mouse and plotted.

Determination of in Vitro Expression on Phospho-S6 Ribosomal Protein (PS6RP)

B16 cells were seeded onto 6-well plates (1.0 × 105 cells/well) and were pre-incubated for 24 h. The medium was changed to NaHCO3-free fresh media at different pH (pH 6.0, 6.2, 6.4, or 7.4) adjusted by adding HCl or NaOH. After 24-h incubation, the cells were lysed with a lysis solution composed of 0.1% Triton X-100, 8 mg/mL NaCl, 8% glycerol, 0.28 mg/mL ethylenediaminetetraacetic acid (EDTA) and 2.42 mg/mL Tris–HCl (pH 7.4) including protease inhibitors (ProteoGuard™, TaKaRa BIO, Shiga, Japan). Total protein contents were measured using DC™ Protein Assay (Bio-Rad Laboratories, CA, U.S.A.). The cell extracts were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) (10 µg total protein/well) using polyacrylamide gels (ATTO, Tokyo, Japan). The expression level of PS6RP in the cells was determined by Western blotting under the following conditions: blocking with 5% bovine serum albumin (BSA) in Tris-buffered saline for 1 h at 37 °C; incubation with a primary antibody of rabbit anti-mouse PS6RP antibody (Cell Signaling Technology, MA, U.S.A.) or rabbit polyclonal antibody to β-Actin (ab16039, Abcam, Cambridge, U.K.) overnight at 4 °C; incubation with a secondary antibody of horseradish peroxidase (HRP)-conjugated anti-rabbit immunoglobulin G (IgG) antibody (Abcam) for 1 h at 37 °C. The expression of PS6RP was visualized with a chemiluminescent substrate (ECL prime, GE Healthcare Bioscience, Buckinghamshire, U.K.) and analyzed with a LAS-4000 system (GE Healthcare Bioscience).

Determination of in Vivo Expression on PS6RP

B16 tumor-bearing mice were orally administered with 7 daily doses of NaHCO3 (500 mg/kg/d) plus MgO (165 mg/kg/d). One day after the last administration, tumors were harvested and snap-frozen in optimal cutting-temperature compound (O.C.T compound, Sakura Finetek Japan, Tokyo, Japan). The frozen tumor sections (7-µm thick) were prepared using a cryostat and mounted on a MAS-coated glass slide (Matsunami Glass, Osaka, Japan). The sections were fixed by incubation in 4% paraformaldehyde for 10 min at room temperature. The tumor sections were stained with a primary antibody of rabbit anti-mouse PS6RP and then with a secondary antibody of Alexa Fluor 488-labeled anti-rabbit antibody (Thermo Fisher Scientific, MA, U.S.A.). The expression of PS6RP was imaged using a BZ-9000 fluorescence microscope (Keyence, Osaka, Japan) with a filter set at Ex. 490 nm and Em. 525 nm. Fluorescent intensity in the region of interest (ROI) was quantified using ImageJ, an open source Java-written program provided by NIH.

Therapeutic Effects of Anti-PD-1 Antibody Combined with Alkalinizing Agents in a Murine Melanoma Model

B16 tumor-bearing mice were orally administered with either NaHCO3 (500 mg/kg/d) plus MgO (165 mg/kg/d) or KNa-cit (500 mg/kg/d) every day. The daily oral administration was performed using a feeding needle and was continued for over 14 d until the mice were terminated at 20% body weight loss. On 3 d after the first administration of alkalinizing agents, in the combination group, the mice were intraperitoneally injected with a dose of anti-PD-1 (5.0 mg/kg/d). The day when the anti-PD-1 was injected was set at Day 0 in the Figures. Volume of tumor and body weight of the treated mice were recorded twice weekly, and survival of the mice was monitored daily. Urinary pH was measured twice weekly using the pH test paper (pH 5.5–9.0, AS ONE). The tumor growth inhibition (%TGI) was calculated using the following formula (TV: Tumor volume).28)

  

The mean survival time (MST) was recorded the mortality on a daily basis, and the increased life span (%ILS) for the treatment groups was calculated using the following formula.29,30)

  

Effects of Combined Anti-PD-1 and KNa-Cit Treatment on the Infiltration of Immune Cells into Tumors

B16 tumor-bearing mice were orally administered, using a feeding needle, with 7 doses of KNa-cit (500 mg/kg/d) daily from Day 0 to Day 6 and were intraperitoneally injected with a single dose of anti-PD-1 antibody (5.0 mg/kg/d) on Day 4. On Day 7, the tumor was harvested, and the tumor cell suspensions were prepared using gentle dissociation with a MACS Dissociator (Miltenyi Biotec, Bergisch Gladbach, Germany) as described previously.31) The tumors were incubated with a mixture of Collagenase Type I (FUJIFILM Wako Pure Chemical Corporation) and Dispase II (Roche Diagnostic, Mannheim, Germany) for 40 min at 37 °C. The suspensions were further dissociated in the presence of deoxyribonuclease (DNase) I (Roche Diagnostic) and were then filtered through a cell strainer (100 µm, Corning). The prepared cells were incubated with the following 4 combinations of antibody. (1) CD8+ T cells: eFluor 660-labeled anti-mouse CD8a antibody (eBioscience, CA, U.S.A.), (2) natural killer (NK) cells: fluorescein isothiocyanate (FITC)-labeled anti-mouse CD49b (Integrin alpha 2) antibody (eBioscience), (3) tumor-associated macrophages (TAM): eFluor 660-labeled anti-mouse F4/80 antibody (eBioscience) and Alexa Fluor 488-labeled anti-mouse CD206 antibody (BioLegend, CA, U.S.A.), (4) regulatory T cells (Treg): eFluor 660-labeled anti-mouse CD4 antibody (eBioscience) and Alexa Fluor 488-labeled anti-mouse CD25 antibody (eBioscience). Flow cytometry analysis was performed using a Gallios flow cytometer (Beckman Coulter, CA, U.S.A.).

Effects of the Treatment with Anti-PD-1 and pHe Condition on Secretion of Interferon-Gamma (IFN-γ) from Spleen Cells

B16 tumor-bearing mice were intraperitoneally injected with a single dose of anti-PD-1 antibody (5.0 mg/kg/d). On 3 d post injection, the spleen was harvested, and the spleen cell suspensions were prepared as described previously.32) Spleen cells were obtained by pressing spleens through a cell strainer (100 µm, Corning) and suspended in RPMI-1640 medium. Red blood cells in the cell suspension were lysed via incubation with ammonium chloride lysis buffer (0.83% NH4Cl) for 3 min. The spleen cells were cultured in 24-well plates (5 × 106 cells/well) in the presence or absence of B16 cells (spleen cells : B16 cells = 10 : 1) using NaHCO3-free RPMI-1640 media at pH 6.2 or 7.4, adjusted by adding HCl or NaOH. After 48-h incubation, the concentration of IFN-γ in the supernatant was determined using a mouse IFN-γ enzyme-linked immunosorbent assay (ELISA) Kit (Proteintech Group, IL, U.S.A.).

Statistical Analysis

Statistical differences between the groups were evaluated by ANOVA with the Tukey post-hoc test using the Prism 8 software (GraphPad Software, San Diego, CA, U.S.A.). All values are reported as the mean ± standard deviation (S.D.). The levels of significance were set at * p < 0.05, ** p < 0.01, *** p < 0.001.

RESULTS

HCO3 Concentrations in Serum and Urine, and Urinary pH, after Oral Administration of an Alkalinizing Agent

Serum HCO3 concentrations, urinary HCO3 concentrations and urinary pH values were investigated in naive mice after a single oral administration with NaHCO3 plus MgO (Fig. 1A). Orally administered MgO proceeds the following reactions in stomach: 2HCl + MgO→MgCl2 + H2O,33) and the resultant MgCl2 absorbs undesired CO2 gas produced by orally administered NaHCO3. Serum HCO3 concentrations increased two-fold up until 6 h post administration and decreased to basal level by 24 h. Urinary HCO3 concentrations increased rapidly within 1 h after administration and then decreased to basal levels by 24 h. Urinary pH increased to over 8.0 within 1 h after administration and then decreased to 6.0–7.0 by 24 h. The urinary pH values were correlated with changes in serum HCO3 concentrations and urinary HCO3 concentrations. The effect of another alkalinizing agent, KNa-cit, was investigated on the same parameters (Fig. 1B). Citrate, a main component of KNa-cit, is converted to HCO3 in a body and helps to correct the acid buildup in the blood.26) A single administration of KNa-cit, given to naive mice, tended to increase serum HCO3 concentrations over 24 h. Urinary HCO3 concentrations increased rapidly by 1 h and then decreased to the base level by 4 h. Urinary pH increased to approx. 7.4 within 1 h after administration and then fell back to 6.0–7.0 by 4 h. These results totally demonstrates that an oral administration of an alkalinizing agent, including NaHCO3 and KNa-cit, expands blood HCO3 concentrations, corresponding to urinary HCO3 concentrations and urinary pH values, and may subsequently increase the tumor pHe.

Fig. 1. Serum HCO3 Concentrations, Urinary HCO3 Concentrations and Urinary pH Values after an Oral Administration of Alkalinizing Agent

BALB/c mice were orally administered with a dose of either (A) NaHCO3 (500 mg/kg) plus MgO (165 mg/kg) or (B) KNa-cit (500 mg/kg). At selected time points post administration (0, 0.5, 1, 2, 4, 6, 8, and 24 h), blood and urine were collected from the treated mice. Serum HCO3 concentrations and urinary HCO3 concentration were measured using the HCO3 measurement kit. Urinary pH values were measured using pH test papers. The data are means ± standard deviation (S.D.) (n = 5–6).

Change in Tumor pHe by Oral Administration of an Alkalinizing Agent

To assess the change of interstitial tumor pHe after treatment with NaHCO3, several approaches were applied. pH electrode is one of adequate tools to directly measure the intratumor pH.20,27) As shown in Fig. 2A, subcutaneous pH in normal mice, as a reference, was neutral level (pH 7.15). The intratumor pH in non-treated tumor-bearing mice was acidified at pH 6.67, which is consistent with the other observations.34) Meanwhile, the oral treatment with NaHCO3 clearly increased the intratumor pH to neutral level at pH 6.90.

Fig. 2. Observation of Tumor pHe after an Oral Administration of Alkalinizing Agent

(A) Colon-26 tumor-bearing mice were allowed to drink water containing 200 mM NaHCO3 ad lib. After 14-d ingestion of NaHCO3 solution, the interstitial pHe in the tumor was measured using microelectrodes with pH meter. A reference electrode or a pH electrode was stabbed into the subcutaneous region or the center of tumor, respectively. One measurement was taken at each mouse and plotted with means (n = 7–9, *** p < 0.001). (B) B16 cells were cultured in 6-well plates (1.0 × 105 cells/well) in media of different pH (pH 6.0, 6.2, 6.4 or 7.4). After 24 h incubation, the expression level of PS6RP and β-actin in the cells was determined by Western blotting. (C) B16 tumor-bearing mice were orally administered with 7 daily doses of NaHCO3 (500 mg/kg/d) plus MgO (165 mg/kg/d). On one day after the last administration, the frozen-tumor sections were prepared. PS6RP in the sections was stained by immunohistochemistry. The localization of PS6RP in both peripheral area and central area in the tumor sections was imaged using a fluorescence microscope. (D) The ROI in the images was quantified using ImageJ. The data are means ± S.D. (n = 5, RFU: relative fluorescence unit, * p < 0.05 vs. control). (Color figure can be accessed in the online version.)

It has been reported that the expression of PS6RP, a downstream target of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway,35) was higher at physiological pHe than at acidic pHe.19) Thus, the expression of PS6RP in tumor cells was studied as a function of tumor interstitial pHe. In vitro PS6RP expression in B16 melanoma cells was increased when cultured in media at physiological pH (pH 7.4) compared to acidic pHs (pH 6.0–6.6) (Fig. 2B). In vivo PS6RP expression in the tumor was then evaluated in a B16 tumor-bearing mouse model. In the peripheral area of untreated tumors, a small somewhat focal increase in PS6RP expression was detected with little activity in the interior (Fig. 2C). In tumors treated with NaHCO3, PS6RP expression was increased both at the tumor periphery and the tumor interior compared to untreated tumors (Figs. 2C, D).

Regarding the treatment with KNa-cit, the effect on neutralizing the B16 tumor was weaker compared to NaHCO3 (data not shown), because KNa-cit could induce the tumor neutralization via producing NaHCO3 that is one metabolite after oral administration of KNa-cit. These results strongly suggest that the treatment with an alkalinizing agent, such as NaHCO3 and KNa-cit, can neutralize the tumor acidified interstitial pHe, presumably owing to the increasing intratumor HCO3 concentrations, corresponding to blood HCO3 concentrations.

Combined Effects of Oral NaHCO3 with Anti-PD-1 on B16 Tumor-Bearing Mouse Model

The effects of increasing tumor pHe by oral NaHCO3 on tumor response of anti-PD-1 were studied in a B16 tumor-bearing mouse model. As shown in Fig. 3A, the treatment with NaHCO3 plus MgO scarcely decrease the growth rate of tumors compared to control (%TGI by 22.3%), and the treatment with anti-PD-1 alone delayed tumor growth (%TGI by 42.1%). The combination of NaHCO3 plus MgO with anti-PD-1 was even more effective in suppressing tumor growth (%TGI by 80.1% > 22.3 + 42.1%), which means that the combination treatment could synergistically enhance the tumor-suppressive effects of anti-PD-1. Survival of the treated mice was also monitored (Fig. 3B) and the %ILS was calculated. The treatment with NaHCO3 plus MgO did not increase the %ILS compared to the untreated control mice. Anti-PD-1 alone, and in combination with NaHCO3 plus MgO, increased the %ILS by 33.7 and 52.6%, respectively. Regarding the body weight changes of the mice (Fig. 3C), there are no remarkable changes in body weight among the all groups, indicating a lack of severe adverse effects. A negative correlation was observed between urinary pH values and tumor volumes at Day 14 in the groups of NaHCO3 plus MgO and combination with anti PD-1 (Fig. 3D); the higher the urinary pH values, the lower the tumor volumes. Survival periods of time were correlated with decreasing urinary pH values in the groups of NaHCO3 plus MgO and the combination treatment (Fig. 3E). As shown in Fig. 1A, oral administration of NaHCO3 plus MgO increased the urinary pH value up to 8.5 to 9.0. Consistently, sequential treatment with NaHCO3 plus MgO or the combination with anti-PD-1 increased the urinary pH up to 8.5 to 9.0 in tumor-bearing mice until Day 11 (Fig. 3E). However, on Day 14, the urinary pH in mice treated with NaHCO3 plus MgO was clearly acidified compared to that in the combination. This would be due to the reason that the tumor in mice treated with NaHCO3 plus MgO substantially grew up to 5000 mm3 in size (Fig. 3A), produced excess H+ and acidified the body. In contrast, the combination treatment of NaHCO3 plus MgO with anti-PD-1 effectively suppressed the growth of tumor at 1300 mm3 in size on Day 14 (Fig. 3A). Under such situation, small tumor produced less H+. and, in turn, the combined treatment still remained the urinary pH high. These results suggest that the treatment with an alkalinizing agent can improve responses to an immune checkpoint inhibitor.

Fig. 3. Combined Treatment with NaHCO3 Plus MgO and Anti-PD-1 in Murine B16 Melanoma

B16 tumor-bearing mice were orally administered with NaHCO3 (500 mg/kg/d) plus MgO (165 mg/kg/d) every day. On Day 3, in the combination group, the mice were intraperitoneally injected with a dose of anti-PD-1 (5.0 mg/kg/d). The day when the anti-PD-1 was administered was set at Day 0 in the figures. (A) Tumor volumes, (B) survival and (C) body weights of the treated mice were monitored. (D, E) Urinary pH was measured using pH test papers. In the treatment groups receiving NaHCO3 plus MgO and combination with anti-PD-1, correlations were plotted (D) between the urinary pH change and tumor volumes on Day 14 and (E) between urinary pH change and survival of the mice. The data in A, C, and pH changes in E are means ± S.D. (n = 8–10, * p < 0.05, *** p < 0.001 vs. control, ##p < 0.01 vs. NaHCO3 plus MgO). (Color figure can be accessed in the online version.)

Combined Effects of Oral KNa-Cit with Anti-PD-1 on B16 Tumor-Bearing Mouse Model

Effects of KNa-cit, another alkalinizing agent, on the therapeutic response of anti-PD-1 was investigated. Treatment with KNa-cit alone did not affect tumor growth rate compared to that in control group (Fig. 4A). The combined treatment with KNa-cit and anti-PD-1 showed the tendency to inhibit the tumor growth (%TGI by 41.7%). In addition, the combined treatment prolonged the survival period of time in treated model mice with good ILS (24.2%) (Fig. 4B), which was consistent with the results of the combined treatment with NaHCO3 plus MgO and anti-PD-1 (Fig. 3B). All the treatment groups showed no severe side effects during the treatment term (Fig. 4C).

Fig. 4. Combined Treatments with KNa-Cit and Anti-PD-1 in Murine B16 Melanoma

B16 tumor-bearing mice were orally administered with KNa-cit (500 mg/kg/d) every day. On Day 3, in the combination group, the mice were intraperitoneally injected with a dose of anti-PD-1 antibody (5.0 mg/kg/d). The day when the anti-PD-1 antibodies was given was set at Day 0 in the figures. (A) Tumor volumes, (B) survival times, and (C) body weights of the treated mice were monitored. The data in A and C are means ± S.D. (n = 7–8, ** p < 0.01 vs. control). (Color figure can be accessed in the online version.)

Infiltration of Immune Cells into B16 Tumors after the Combined Treatment with an Alkalinizing Agent and Anti-PD-1

Pilon-Thomas et al. reported that increasing of tumor pHe enhances the infiltration of immune cells into tumor tissues.17) In the current study, we experimentally confirmed this observation using KNa-cit, because KNa-cit could produce much comprehensive alkalinizing effects via producing a metabolite of NaHCO3 and shifting electrical equilibrium in a body. As shown in Fig. 5A, treatment with anti-PD-1 alone increased the tumor-infiltration of CD8+ T cell, an important cytotoxic effector cell type in anti-tumor immunity.34) Combined treatment with KNa-cit and anti-PD-1 further increased the infiltration of CD8+ T cell into tumors to a small degree. Treatment with anti-PD-1 alone tended to increase the population of TAM, M2-phenotype macrophages that promote the proliferation and survival of tumor cells,36) but the combined treatment with KNa-cit and anti-PD-1 had little effect. Change in the infiltrations of NK cells and Treg was not observed in this study. Treatment with KNa-cit alone did not affect the tumor infiltration of immune cells including CD8+ T cell, TAM, NK cell, and Treg (Fig. 5B).

Fig. 5. Infiltration of Immune Cells into the B16 Tumors in Combination Treatment with an Alkalinizing Agent and Anti-PD-1

B16 tumor-bearing mice were orally administered with KNa-cit (500 mg/kg/d) daily from Day 0 to Day 6 and were intraperitoneally injected with a dose of anti-PD-1 (5.0 mg/kg/d) on Day 4. On Day 7, tumors were harvested. (A) Tumor-infiltrating immune cells (CD8+ T cell, TAM, NK cell, and Treg) after the treatment with anti-PD-1 or the combination of anti-PD-1 with KNa-cit were determined by flow cytometry and are shown as the percentage of total tumor cells. (B) Tumor-infiltrating immune cells after the treatment with KNa-cit alone were determined by flow cytometry. (C) The spleen was harvested from the same mice treated with/without anti-PD-1, and the spleen cells were cultured in 24-well plates (5 × 106 cells/well) in the presence or absence of B16 cells under different pH conditions (pH 6.2 or 7.4). After 48 h incubation, secreted IFN-γ in the media was determined using a mouse IFN-γ ELISA Kit. The data are means ± S.D. (n = 4, ** p < 0.01, *** p < 0.001, n.s.: not significant). (Color figure can be accessed in the online version.)

We also elucidated the activation of immune cells under our treatment conditions by studying the ex vivo secretion of the IFN-γ cytokine from spleen cells collected from tumor-bearing mice treated with/without anti-PD-1 (Fig. 5C). When co-cultured with B16 cells, the spleen cells secreted higher levels of IFN-γ at physiological pH condition (pH 7.4) compared to acidic pH condition (pH 6.2) in both treatment groups. In addition, at pH 7.4, spleen cells from control tumor-bearing mice produce higher levels of IFN-γ compared to those from the anti-PD-1-treated mice, which might depend on the exhaustion of splenic T cells by anti-PD-1 immunotherapy.37) Spleen cells, in the absence of B16 cells, produced little or no IFN-γ (Fig. 5C). The ex vivo results indicate that the combined treatment with an alkalinizing agent and anti-PD-1 somewhat enhances the infiltration of cytotoxic CD8+ T cells into the tumor and would activate the infiltrated immune cells via increasing tumor interstitial pHe by oral alkalinizing agent.

DISCUSSION

Recent clinical reports suggest that acid-base balances in vivo are important for patient health status and may be involved in the clinical outcomes. Chan et al. reported that, in colorectal cancer patients undergoing resection of their primary tumors, the group with low serum HCO3 concentrations had a significantly lower 30-d overall survival than the group with normal HCO3 levels.38) In addition, Hamaguchi et al. reported that, in advanced pancreas cancer patients treated with an alkalinizing therapy (an alkaline diet with supplementary oral NaHCO3), the median overall survival from the start of alkalinizing therapy of the patients with high urinary pH (>7.0) was significantly longer than those with low urinary pH (≤7.0) (16.1 vs. 4.7 months; p < 0.05).39) In this study, we confirmed that the treatment with either NaHCO3 or KNa-cit increases serum HCO3 concentrations, as predicted by increase of urinary pH values (Fig. 1), and consequently neutralize the tumor interstitial pHe at both peripheral and interior regions (Fig. 2). These suggest that the alkalinizing treatment can improve the malignant tumor microenvironment, such as acidic nature, via increasing serum HCO3 concentrations.

It is reported that tumor acidity can influence responses to immunotherapy and ionizable anticancer drugs.1416) Acidification of the pHe may promote escape of solid tumors from immune surveillance and potentially limit the efficacy of immunotherapies such as an immune checkpoint inhibitor.17,40) One can hypothesize that therapeutic interventions designed to increase the pHe in patients may be able to improve therapeutic outcomes. We tested this hypothesis in vitro and in vivo in murine tumor models and confirmed that increasing tumor pHe with an oral alkalinizing agent, NaHCO3 plus MgO or KNa-cit, increased the response to anti-PD-1 in a B16 tumor-bearing mouse model (Figs. 3, 4), and there was a correlation between urinary pH values and the antitumor effects of anti-PD-1 immunotherapy (Fig. 3). To the best of our knowledge, this is the first report to suggest that the treatment with an oral alkalinizing agent can improve the tumor-growth suppressive effects of an immune checkpoint inhibitor via elevating serum HCO3 concentrations and subsequent neutralizing the acidic tumor interstitial pHe.

In the current study, we showed that the treatments with an alkalinizing agent, either NaHCO3 plus MgO or KNa-cit, enhanced the therapeutic effect of anti-PD-1 immunotherapy, which is consistent with the previous results.17) The increased responses were hypothesized to be a result of an alkalinizing agent having the effect of increasing tumor pHe and enhancing T-cell infiltration into tumors. Acidic pHe is reported to suppress not only growth of tumor cells,41) but also several immune functions relating to immune checkpoint therapy.40) In vitro studies have shown that T-cell functions, including the interleukin (IL)-2 secretion and T-cell receptor activation, were suppressed in acidic pHe conditions.42) Our current study demonstrated that the combined treatment with KNa-cit and anti-PD-1 increased the infiltration of CD8+ T cells into the tumor (Fig. 5A) and could activate the cytotoxic immune cells (Fig. 5C). It has been reported that acidic pHe resulted in an increase in the numbers of immune-suppressor cells such as myeloid-derived suppressor cells (MDSC), Treg, and TAM, switching the immune cytokine profile from a cytotoxic T-helper 1 (Th1) phenotype to an immunosuppressed Th2 phenotype.43) Therefore, tumor interstitial acidification could suppress tumor-specific immune responses and increase tumor growth. We observed that combined treatment with KNa-cit and anti-PD-1 decreased the immune suppressive TAM population in the tumor (Fig. 5A), corresponding to serum HCO3 concentrations (Fig. 1) and neutralization in tumor pHe (Fig. 2), which would be expected to enhance the therapeutic effect of anti-PD-1. It would appear that increasing tumor pHe by an oral alkalinizing agent can restore T-cell functions including IFN-γ abrogation and tumor necrosis factor (TNF)-α secretion.17) This could be one explanation for the improved responses that we observed for immunotherapy with anti-PD-1 in combination with an alkalinizing agent.

It has been assumed that, after oral administration, NaHCO3 plus MgO and KNa-cit produce HCO3 via the following mechanism: the gastrointestinal pH is increased by the buffering effect of NaHCO3, enhancing secretion of H+ ions to maintain homeostasis in the gastrointestinal pH and with the production of HCO3. HCO3, produced from NaHCO3, is absorbed from the intestinal tract, and serum HCO3 concentration gradually increases (Fig. 1A). KNa-cit is absorbed from the intestinal tract into blood and dissociated into its constituent ions. The citrate anion is excreted through the urine, causing a shift in the electrical equilibrium.44,45) In order to recover this homeostasis, serum HCO3 concentrations increase accompanied with a decrease of serum H+ ions,26) corresponding to decreasing H+ ions in tumor tissue. Although the mechanisms producing HCO3 are different, the rates of HCO3 appearing in, or disappearing from, the blood were similar for both alkalinizing agents (Fig. 1). It is well known that excess HCO3 in the blood is eliminated through glomerular filtration into urine; renal filtration regulates blood levels of HCO3 and acid secretion.24) Accordingly, urinary HCO3 concentrations and pH values were increased in conjunction with increases in serum HCO3 concentrations. Interestingly, in KNa-cit, urinary pH corresponding to serum HCO3 concentrations rapidly dropped to base levels compared to NaHCO3 plus MgO (Fig. 1B). This might be due to the presence of excess citrate ions secreted in urine that would tend to acidify the urine. Nevertheless, both of the alkalinizing agents that we used in this study would elevate serum HCO3 concentrations, which may have been sufficient to elevate tumor pHe.

In conclusion, we demonstrated that the treatment with an alkalinizing agent increases the acidic tumor interstitial pHe via elevating serum HCO3 concentrations, corresponding to urinary HCO3 concentrations and urinary pH values. Our results imply that serum HCO3 concentrations, corresponding to tumor pHe, and urinary pH values may be important factors to predict clinical outcomes of some types of anticancer treatments combined with an alkalinizing agent.

Acknowledgments

The authors are grateful to Professor Emerita Theresa M. Allen, University of Alberta for her helpful advice in developing the English manuscript.

Conflict of Interest

K.E. is President of Delta-Fly Pharma, Inc. The other authors declare no conflict of interest.

REFERENCES
 
© 2021 The Pharmaceutical Society of Japan
feedback
Top