Genotoxic stress induces cellular senescence and PDK4 expression
Pyruvate enters the TCA cycle through PDH, while PDK molecules (PDK1-4) inhibit PDH activity and promotes the switch from mitochondrial oxidation to cytoplasmic glycolysis. PDK4 is located in the mitochondrial matrix and inhibits the PDH complex by phosphorylating its E1α subunit, thereby contributing to glucose metabolism regulation (Thapa et al., 2018). Although predominantly expressed in the muscle and affects the metabolic fate of glucose during exercise, PDK4 is intensively studied in multiple cancer types (Leclerc et al., 2017; Sun et al., 2017; Sun et al., 2014). However, insights into PDK4 expression in normal tissue microenvironments and its inducibility in response to stressful insults remain limited. We recently noticed that the stromal cell line PSC27, which is of human prostate origin and comprising mainly fibroblasts but with a minor percentage of non-fibroblast cell lineages such as endothelial cells and smooth muscle cells, produces a large array of SASP factors upon exposure to cytotoxic insults specifically genotoxic chemotherapy or ionizing radiation (Sun et al., 2012; Zhang et al., 2018). Interestingly, PDK4 emerged as one of the upregulated factors, together with a list of typical SASP components, as previously revealed by our microarray profiling (Fig. 1a and Supplementary Fig. 1a) (Sun et al., 2012). To confirm the finding, we expanded by employing several alternative approaches to induce senescence, including replicative exhaustion (RS), overexpression of p16INK4a (p16) and HRasG12V (RAS), respectively. These treatment caused comprehensive cellular senescence, with an efficacy resembling that of DNA-damaging agents such as radiation (RAD), bleomycin (BLEO) and hydrogen peroxide (HP) (SA-β-Gal positivity and BrdU incorporation, Supplementary Fig. 1b-d). In each case, we observed a significant PDK4 induction (Fig. 1b-c).
Expression analysis of several cell lines of human prostate origin suggested that stromal cells are indeed more PDK4-inducible than cancer epithelial cells, implying a special mechanism that supports PDK4 production in prostate stromal cells posttreatment (Fig. 1d). Data from several additional fibroblast lines consistently supported a robust induction of PDK4 upon genotoxic treatment by anticancer agents (Fig. 1e). Notably, the transcript expression pattern of PDK4 largely phenocopied that of a group of hallmark SASP factors including MMP1, WNT16B, SFRP2, SPINK1, MMP3, CXCL8, EREG, ANGPTL4 and AREG, which exhibited a gradual increment until entering a platform within 7-8 days after treatment (Fig. 1f-g). Among the human PDK family (PDK1-4 isozymes), PDK4 appeared to be the only member that is readily inducible by genotoxic stress, with a tendency similar to that of CXCL8, an index of the SASP expression (Fig. 1h-i).
PDK4 expression in stroma predicts adverse clinical outcomes post-chemotherapy
Experimental data derived from in vitro assays prompted us to further determine whether PDK4 induction occurs within the tumor microenvironment (TME), a pathological entity where a plenty of benign stromal cells reside. We first chose to analyze clinical samples of a cohort of prostate cancer (PCa) patients who developed primary tumors in the prostate and underwent chemotherapeutic regimen involving genotoxic agents such as mitoxantrone (MIT). Surprisingly, PDK4 was found markedly expressed in prostate tissues of these patients after chemotherapy, but not before (Fig. 2a). Basically in line with our in vitro data, upregulated PDK4 was generally localized in the stroma, in a sharp contrast to the adjacent cancer epithelium which had limited or no staining.
PDK4 production in patient samples pre- versus post-chemotherapy was quantitatively measured by a pre-established pathological assessment procedure, which allowed precise evaluation of the expression of a target protein per immunohistochemistry (IHC) staining intensity (Fig. 2b). Transcript analysis upon laser capture microdissection (LCM) of cell lineages from primary tissues suggested that PDK4 was more readily induced in the stromal rather than cancer cell subpopulations (P < 0.0001 versus P > 0.05) (Fig. 2c). To substantiate PDK4 inducibility in vivo, we profiled a subset of PCa patients whose pre- and post-chemotherapy biospecimens were both accessible, and found remarkably upregulated PDK4 in the stroma, but not cancer epithelium, of each individual post-chemotherapy (Fig. 2d-e). We noticed the dynamics of PDK4 expression in the damaged TME largely in parallel to that of CXCL8 and WNT16B, two canonical SASP components (Fig. 2f). Expression pattern of these factors were largely consistent with that of senescence markers including p16INK4a and p21CIP1 in tumor foci, suggesting an inherent correlation of PDK4 induction with cellular senescence and the SASP (Fig. 2f). Of note, Kaplan-Meier analysis of PCa patients stratified according to PDK4 expression in tumor stroma suggested a significant but negative correlation between PDK4 protein level and disease-free survival (DFS) in the treated cohort (P < 0.05, log-rank test) (Fig. 2g).
The distinct pathological properties of PDK4 in PCa were subsequently reproduced by an extended study, which was designed to recruit clinical cohorts of human breast cancer (BCa) patients (Supplementary Fig. 2a-d). Implicating the functional roles of PDK4, such as working as a critical regulator of epithelial-to-mesenchymal transition (EMT) and drug resistance of human cancers (Sun et al., 2014), data from gene expression profiling interactive analysis (GEPIA) with the cancer genome atlas (TCGA) and genotype-tissue expression (GTEx) databases indicated that PDK4 expression in cancer cells per se is associated with the poor prognosis of some, but not all cancer types (Supplementary Fig. 2e-f). Thereby, in contrast to former studies that mainly focused on PDK4 expression in cancer cells per se, our data consistently suggest that PDK4 induction in tumor stroma may act as an SASP-associated independent predictor of clinical prognosis, holding the potential to be exploited for stratifying the risk of disease relapse and clinical mortality of posttreatment patients. Given such a pathological relevance, it is reasonable to speculate that PDK4 production by the stroma may have a causal role in senescence-related conditions, such as cancer progression.
Senescent cells exhibit a distinct profile of glucose metabolism
A typical feature of cancer cells is the ability of reprogramming energy metabolism to fuel their expansion and survival, while enhanced mitochondrial function plays important roles in tumor development (An and Duan, 2022). One of the major hallmarks of senescent cells is that they remain metabolically active and synthesize a plethora of protein factors (SASP) with a capacity to affect other cells of the host microenvironment locally or systemically (Prasanna et al., 2021). Former studies on the metabolism of cellular senescence demonstrated that levels of both glucose consumption and lactate production are elevated during senescence (Calcinotto et al., 2019). While increased expression of glucose transporter and glycolytic enzymes during cellular senescence was observed, to date relevant data were mostly derived from cancers such as lymphomas and melanomas, or senescent cells induced by activation of oncogenes such as BRAFV600E (Dorr et al., 2013; Kaplon et al., 2013). In contrast, the metabolic feature of glucose, a major energy source of senescent cells, and the influence of such a metabolic profile on surrounding tissue homeostasis, remain largely underexplored and merits in-depth understanding.
Glucose is the primary carbon source to the tricarboxylic acid (TCA) cycle, followed by glutamate and aspartate (non-protonatable amino acids as glutamine or asparagine, respectively) as secondary sources (Fig. 3a) (Kirsch et al., 2021). We first interrogated the metabolic pattern of glucose upon uptake by senescent cells, as glucose is supposed to act as a principal contributor to the TCA cycle when cells enter senescence, a stage that is considered metabolically active (Pan et al., 2022). Experimental data from assessment of mitochondrial dynamics and cellular bioenergetics with the XF24 Extracellular Flux analyzer (Seahorse bioanalyzer) indicated significantly elevated glycolytic activity in senescent human stromal cells, as reflected by enhanced production of metabolites including but not limited to dihydroxyacetone phosphate (DHAP), glyceraldehyde-3-phosphate (GAP) and 3-phosphoglycerate (3-PG) (Fig. 3b). Increased levels of GAP and 3-PG imply further utilization of a number of middle metabolites, such as citrate, α-ketoglutarate, glutamate, succinate, fumarate and malate in TCA cycle, which was indeed substantiated by data from metabolic profiling with the Seahorse bioanalyzer (Fig. 3c and Supplementary Fig. 3a-f). Thus, bioactivities of both glycolysis and TCA cycle were significantly enhanced in senescent cells, as reflected by an overall profiling with assays of stable isotope labelling with a uniformly labelled U-13C6 glucose tracer and fractioning of metabolites derived from labelled glucose (Fig. 3d). We further noticed that these metabolic changes were accompanied by remarkable perturbations in mitochondrial ultrastructure of senescent cells, particularly the enlarged size and abnormal shape as revealed by transmission electron microscopy (TEM), a phenomenon indicative of ultrastructural damage of mitochondria and suggesting potential mitochondrial dysfunction associated with oxidative stress upon cellular senescence (Fig. 3e). These observations are largely consistent with former studies regarding abnormal mitochondrial phenotypes including their mass, dynamics and structure in senescent cells (Martini and Passos, 2022).
We next measured the levels of extracellular fluids. Strikingly, the amounts of both pyruvate and lactate released to the extracellular space were considerably enhanced in senescent cells relative to their control counterparts (Fig. 3f). These changes were accompanied by alterations in oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), suggesting elevated metabolic activities associated with glucose utilization (Fig. 3g and Supplementary Fig. 3g-i). Correspondingly, we observed elevated ATP production, basal respiration, maximum respiration in senescent cells, a tendency indicative of tight connection of TCA cycle and oxidative phosphorylation (OXPHOS) but further promoted when PDK4-IN-1, an anthraquinone derivative and a potent inhibitor of PDK4 (referred to as PDK-IN hereafter) (Lee et al., 2019), was applied to culture (Fig. 3h-j). However, treatment with PDK4-IN reversed the tendency of such changes in non-mitochondrial oxygen consumption, pH fluctuation, lactate production and H+ (proton) leak, with the overall metabolic data validated by principal component analysis (PCA) scores (PC1 vs PC2) (Fig. 3k-n and Supplementary Fig. 3j). There alterations occurred in parallel with expression changes of glucose uptake-associated molecules and metabolism-related enzymes including glucose transporter 1 (GLUT1), hexokinase 2 (HK2), lactate dehydrogenase A (LDHA), isocitrate dehydrogenase 2 (IDH2), isocitrate dehydrogenase 3 (IDH2), oxoglutarate dehydrogenase (OGDH) and citrate synthase (CS) (Supplementary Fig. 3k). Among them, HK2 and LDHA are glycolysis-related factors, while IDH2, IDH3, OGDH and CS are TCA cycle-associated enzymes. As overexpression of PDK4 per se caused neither cellular senescence nor the SASP (Supplementary Fig. 3l, m), we reasoned that the metabolic profile of senescent cells was correlated with and likely underpinned by expression of key factors involved in glucose consumption and linked with production of pyruvate, lactate and multiple other metabolites. Of note, elevated levels of glycolysis and oxidative phosphorylation were simultaneously observed, suggesting essentially reprogrammed glucose metabolism upon cellular senescence.
Former studies reported that in contrast to proliferating cells, senescent cells exhibit increased glucose transporter and glycolytic enzyme expression levels after chemotherapeutic treatment (Dorr et al., 2013), a tendency largely confirmed by our experimental data (Supplementary Fig. 3k). Steady state glucose concentrations tend to be higher in senescent cells as compared to their control, suggesting an elevated glucose avidity upon senescence. These findings are essentially confirmed by our metabolomics profiling, which underscores the global catabolic nature of senescence-associated metabolic alterations. Taken together, senescent cells develop a distinctive hypermetabolic phenotype characterized of enhanced glycolysis, TCA activity and ATP-boosting oxidative phosphorylation. Increased energy production is a common denominator of senescent cells, which exhibit a specific utilization of energy-generating metabolic pathways, a phenomenon largely reminiscent of the ‘Warburg effect’ typically observed in cancer cells capable of performing a non-oxidative breakdown of glucose (Dorr et al., 2013).
Senescent cells shapes the formation of an acidic microenvironment via PDK4
Data from previous studies indicated that senescent cells are in a hypermetabolic status, more specifically, display a hypercatabolic nature (Dorr et al., 2013), thus promoting us to interrogate whether these cells have a glucose uptake capacity distinct from that of proliferating cells. To address this, we performed another set of metabolic assays. Not surprisingly, a significant increase of glucose uptake by senescent cells was observed, although with the tendency preferentially detected upon TIS (Fig. 4a). Further, the pH of conditioned media (CM) from senescent cells was markedly decreased, a property that again seemed to be more dramatic for cellular senescence induced by genotoxic agents (Fig. 4b). Given the results indicative of an elevated acidification potential as revealed by the ECAR assay (Supplementary Fig. 3g), we reasonably speculated the extracellular formation of an acidic microenvironment by senescent cells, whose metabolism appeared to be distinctly reprogrammed and was characterized with increased secretion of acidic metabolites. Our data suggested that senescent cells can generate an increased amount of lactate, which was higher than their cycling control counterparts (Supplementary Fig. 4a). Multiple studies reported that cancer cells have increased lactate production, OCR level and ATP production, a series of metabolic changes correlated with enhanced glycolysis (Yu et al., 2022; Zhang et al., 2021). Strikingly, we found that many relevant activities of senescent cells were even higher than those of cancer cells selected as of the same organ origin (herein, prostate), such as PC3 and DU145, although with several key features manifesting changes evidently opposite to those of examined cancer lines (Supplementary Fig. 4a-f).
PDK4 is a key enzyme involved in the regulation of glucose and fatty acid metabolism as well as tissue homeostasis, while its overexpression inactivates the PDH complex by phosphorylating the targets and contributes to metabolic flexibility. We assessed the influence of PDK4 expression by transducing a PDK4 construct to human stromal cells, and noticed significantly altered metabolic profile including glucose uptake, lactate production and triglyceride (TG) production, although these changes were largely reversed upon genetic eliminated of PDK4 (Supplementary Fig. 4g-i). Of note, a decreased pH of the CM was observed in the case of PDK4 overexpression in proliferating cells, but subject to counteraction by PDK4 suppression (Supplementary Fig. 4j). We further measured these parameters with senescent cells induced by BLEO treatment, and found markedly increased glucose uptake, lactate production and TG production, but reduced pH of the CM (Supplementary Fig. 4k-n). However, almost all these metabolic changes were substantially reversed, when PDK4 was depleted from PSC27, except the TG levels, a case suggesting a PDK4-mediated antagonism against TG synthesis throughout the TCA cycle in senescent cells (Supplementary Fig. 4k-n). We noticed some factors functionally supporting glycolysis and TCA, including GLUT1, MCT4, HIF1α, PGK1, PGI, CS, IDH2, IDH3A and IDH3B were concurrently upregulated upon TIS, further indicating an overall enhancement of cell metabolism with the glucose as an energy source (Supplementary Fig. 4o).
NAD+ and its reduced form, NADH, are pivotal coenzymes for redox reactions and play critical roles in energy metabolism (Zhao et al., 2018). The intracellular level of NAD+ is frequently altered during aging and upon age-related pathologies. We previously generated SoNar, an intensely fluorescent, rapidly responsive, pH-resistant and genetically encoded sensor for tracking subtle changes in cytosolic NAD+ and NADH redox states by imaging and quantifying the NAD+/NADH ratio in living cells and in vivo (Zhao et al., 2015), but the metabolic profile NAD+ and NADH in senescent cells remains largely undefined (Fig. 4c). We first measured the intracellular NAD+/NADH redox state of PSC27 cells utilizing SoNar’s fluorescence (Fig. 4d-f). The data indicated a remarkably lower NAD+/NADH ratio upon TIS (evidenced by increased NADH/NAD+), a tendency that was essentially subject to reversal by the PDK4 inhibitor, suggesting a remarkably elevated reduction of NAD+ to NADH, a process accompanied by enhanced glycolysis (Fig. 4g, h). As another technical advancement, we designed FiLa, a highly responsive, ratiometric and genetically encoded lactate sensor to monitor the production and consumption of lactate at subcellular resolution (Fig. 4i-j). We observed a remarkable increase in cytosolic lactate upon cellular senescence, which can be abrogated in the case of PDK4 suppression (Fig. 4k-l). The results from fluorescence sensors suggest that the lactate production level increases in parallel to the NAD+/NADH ratio decrease in senescent cells, while both changes are correlated with PDK4 activity. The data not only disclose the concurrent fluctuation of NAD+/NADH conversion and lactate generation, but further substantiate the central role of PDK4 in orchestrating a metabolic profile specifically associated with the occurrence of cellular senescence.
Stromal cells expressing PDK4 alter the expression profile and phenotypes of cancer cells via HTR2B
We next sought to determine the influence of stromal cells expressing PDK4 on their surrounding microenvironment, specifically cancer cells. Since PSC27 originally derived from human prostate, we first chose to examine PCa cells. PSC27-derived CM were employed to treat PCa cells in culture, with cancer cells subject to genome-wide analysis. Data from RNA sequencing (RNA-seq) indicated that 4188 transcripts were significantly upregulated or downregulated (fold change > 2, P < 0.05) in PC3 cells, with 4860 and 3756 transcripts changed in DU145 and M12 cells, respectively (Fig. 5a). We noticed remarkable and comprehensive changes in the biological processes of PCa cells, as evidenced by typically affected activities in signal transduction, cell communication, intracellular transport, energy pathways and metabolism regulation (Fig. 5b and Supplementary Fig. 5a, b). Together, the data suggest a salient capacity of PDK4-expressing stromal cells in reprogramming the transcriptome of recipient cancer cells through production of the CM, the latter of a typically acidic nature.
Among the transcripts significantly upregulated by the CM of PSC27 cells (P < 0.05, FDR < 0.01, top 1000 shown per PCa line, Supplementary Table 1), we noticed that there were 7 transcripts showing up and commonly expressed by PC3, DU145 and M12 cells (fold change > 4, P < 0.01) (Fig. 5c, d). We then chose to focus on a specific gene (or more accurately, transcript), which potentially contributes to malignant alterations of cancer cells. Immunoblots substantiated that expression of 5-hydroxytryptamine receptor 2B (HTR2B), a gene encoding one of the several different receptors for serotonin which belongs to the G-protein coupled receptor 1 family, was markedly induced upon exposure of PCa cells to the CM of PDK4-expressing stromal cells (Fig. 5e). Serotonin is a biogenic hormone that functions as a neurotransmitter and a mitogen, while serotonin receptors mediate the central and peripheral physiologic functions of serotonin, including regulation of cardiovascular functions and impulsive behavior. We interrogated the implications of HTR2B in phenotypic changes of individual PCa cell lines. Of note, the capacities of proliferation, migration and invasion of PCa cells were comprehensively enhanced, as evidenced by our in vitro assays (Fig. 5f-h). More importantly, transduction of HTR2B enhanced the resistance of PCa cells to MIT, a DNA-targeting chemotherapeutic agent administered to cancer patients including those developing PCa (Dueck et al., 2020; Li et al., 2021) (Fig. 5i). The survival curves of cancer cells under genotoxic stress of MIT displayed a shift toward higher concentrations of this drug, as exemplified by the case of PC3 (Fig. 5j). Notably, the CM derived from senescent stromal cells generated by BLEO treatment (PSC27 (BLEO) CM) caused a more dramatic increase of chemoresistance than HTR2B per se, suggesting the presence and contribution of other molecules in the CM of senescent stromal cells, particularly a large number of soluble factors encoded by the full-spectrum of the SASP. However, these gain of functions were generally lost when LY 266097 (Cathala et al., 2020), a selective antagonist of HTR2B, was applied to the culture, suggesting that enhanced malignancies of cancer cells were mainly attributed to the activity of HTR2B after ectopically expressed in these lines (Fig. 5f-j).
As HTR2B is one of the most upregulated genes observed in PCa lines we examined upon treatment with PDK4+ stromal cell CM, whether or not it accounts for the principal force driving malignant changes of recipient cancer cells remain unknown. We then used LY 266097 or gene-specific small hairpin RNAs (shRNAs) to target HTR2B in individual PCa cell lines before performing phenotypic assays. Interesting, the gain of functions conferred by the CM of PDK4+ stromal cells was substantially abrogated in the absence of HTR2B or upon LY 266097 treatment (Supplementary Fig. 5c-g). Thus, HTR2B is a competent factor that mediates the influence of the acidic microenvironment generated by PDK4-expressing stromal cells on recipient cancer cells, while elimination of HTR2B or blockade of HTR2B signaling in cancer cells holds the potential to remarkably weaken their malignant phenotypes.
Therapeutically targeting PDK4 improves chemotherapeutic outcome in preclinical trials
Given the acidic extracellular microenvironment formed by stromal cells expressing PDK4 and its effects of on the biological phenotype and expression profile of cancer cells in vitro, we were tempted to query the pathological consequences of PDK4 induction in the TME under in vivo conditions. To this end, we constructed tissue recombinants by admixing PSC27 sublines with PC3 cells at a pre-optimized ratio of 1:4 before subcutaneous implantation to the hind flank of experimental mice with severe combined immunodeficiency (SCID). The animals were gauged for tumor size at the end of an 8-week period. Compared with tumors comprising PC3 and PSC27Vector, xenografts composed of PC3 and PSC27PDK4 displayed significantly increased sizes (P < 0.01) (Supplementary Fig. 6a). Conversely, PDK4 knockdown (by shRNA) from these PSC27PDK4 cells prior to xenograft implantation markedly reduced tumor volumes (P < 0.01 and P < 0.05, respectively).
To closely mimic clinical conditions involving chemotherapeutic agents, we designed a preclinical regimen which incorporates a genotoxic drug (MIT) and/or the PDK4 inhibitor (PDK4-IN) (Fig. 6a). Two weeks after cell implantation when stable uptake of tumors by host animals were generally observed, a single dose of MIT or placebo was administered at the 1st day of 3rd, 5th and 7th week until the end of the 8-week regimen (Supplementary Fig. 6b). Although PDK4-IN administration did not provide noticeable benefits, MIT treatment caused remarkable tumor shrinkage (57.5% volume reduction), validating the efficacy of MIT as a cytotoxic agent (Fig. 6b). Importantly, when PDK4-IN was combined with MIT, a further decline of tumor volume was observed (35.5%), resulting a total shrinkage by 72.6% as compared with the vehicle control (Fig. 7b).
Not surprisingly, we observed a considerable upregulation of typical SASP factors such as IL6, CXCL8, MMP3, SPINK1 and AREG, accompanied by expression of typical senescence markers including p16INK4a and p21CIP1 in stromal cells of xenografts composed of PC3/PSC27 cells, implying development of an in vivo senescence and expression of the SASP upon MIT treatment (Fig. 6c and Supplementary Fig. 6c). However, PDK4-IN neither induced nor affected cellular senescence and the SASP, as evidenced by results from its administration as a mono agent or combined with MIT (Fig. 6c and Supplementary Fig. 6c). Although senescence was induced in cancer cells in animals undergoing MIT treatment as suggested by p16INK4a and p21CIP1 expression, we did not notice a typical and full-spectrum SASP in these cells of epithelial origin, results largely consistent with findings of our former studies (Chen et al., 2018; Xu et al., 2019). Of note, PDK4 expression was remarkably induced in stromal cell populations, but not in their epithelial counterparts (Supplementary Fig. 6c), basically in line with our in vitro data (Fig. 1d, e). Histological staining indicated elevated SA-β-Gal positivity in tumor tissues of mice that experienced MIT or MIT/PDK4-IN treatment, confirming comprehensive senescence occurrence in these groups (Fig. 6d, e). In contrast, treatment with PDK4-IN itself did not seem to cause induction or suppression of senescence, thus congruent with the nature of this agent, which typically does not target DNA nor damage macromolecules (Fig. 6c-e).
Given the primary in vivo expression results derived from treatments involving MIT and/or PDK4-IN, we next asked how pharmacologically targeting PDK4 could enhance the therapeutic response of tumors. To disclose the possible mechanism(s), we chose to dissect tumors from animals 7 days after initiation of treatment, a timepoint right prior to the development of resistant colonies. In contrast to vehicle, MIT per se caused significant DNA damage and apoptosis in cancer cells (Fig. 6f). Although PDK4-IN alone did not cause typical DDR or cell apoptosis, it showed prominent efficacy in enhancing these therapeutic indices upon combination with MIT (P < 0.05). IHC staining disclosed increased caspase 3 cleavage, a canonical apoptosis indicator, upon MIT administration, a tendency that was further enhanced in the presence of PDK4-IN (Fig. 6g).
To expand, we used LNCaP, a second PCa cell line which expresses androgen receptor (AR) and is routinely employed as a hormone-responsive cell model. To produce an AR-naïve setting, we circumvented experimental castration, but followed the same protocol designed for PC3-tailored therapeutic cohorts. We noticed significantly reduced volumes of LNCaP/PSC27 tumors when mice underwent MIT/PDK4-IN co-treatment, in contrast to MIT administration only (36.1%) (Supplementary Fig. 6d). Similar results were observed when 22Rv1, a castration-resistant PCa cell line, was applied to replace LNCaP for in vivo assays (35.3%) (Supplementary Fig. 6e). As supporting efforts to exclude the possibility of tumor type specificity, we generated tumors composed of A549, a non-small cell lung cancer (NSCLC) line, and HFL1, a lung fibroblast line. The results largely produced those observed in animals carrying tumors developed from PCa lines (34.0%) (Supplementary Fig. 6f). Together, these data suggest that specific targeting of PDK4, a kinase responsible for lactate production during glucose metabolism and formation of an acidic microenvironment, specifically in a treatment-damaged TME which harbors a number of senescent cells, can substantially promote tumor regression in chemotherapeutic settings, a process independent of androgen regulation or AR signaling of prostate tumors per se. We hereby conclude that the resistance-minimizing effects of PDK4-targeting strategy are not limited to a specific cancer type, but may have implications to a wide range of malignancies.
We next assessed tumor progression consequence by comparing the survival of different animal groups in a time-extended preclinical cohort, with PCa mice as a pilot model. In the course of tumor growth monitoring, a bulky disease was considered developing once the tumor burden was prominent (size ≥ 2000 mm3), an approach described previously (Melisi et al., 2011; Zhang et al., 2018). Mice that received MIT/PDK4-IN combinational treatment displayed the most prolonged median survival, gaining a 40.9% longer survival when compared with those treated by MIT only (Fig. 6h, green vs blue). However, PDK4-IN treatment alone did not achieve significant benefits, as it conferred only marginal survival advantage (Fig. 6h, brown vs red). Thus, targeting PDK4 in the TME affects neither tumor growth nor animal survival, while MIT/PDK4-IN co-treatment has the competence to significantly improve both parameters.
Importantly, to establish the safety and feasibility of such a therapeutic regimens, we conducted routine pathophysiological appraisal. The data supported that either single or combinatorial treatment was well tolerated, as evidenced by body weight maintenance throughout the therapeutic timeframe (Supplementary Fig. 7a). Further, there were no significant perturbations in the serum level of creatinine, urea and metabolic activities of liver enzymes (ALP and ALT) (Supplementary Fig. 7b). Additional data from mice developing NSCLC carcinomas and treated by DOX/PDK4-IN, or DOX/PDK4-IN-treated immunocompetent animals (C57BL/6J background), generally phenocopied PCa animals by manifesting no routine blood count fluctuations, thus further validating these findings (Supplementary Fig. 7c-f). Our data support that strategies combining a PDK4-targeting agent with classical chemotherapy hold the potential to enhance tumor responses without causing severe systemic cytotoxicity.
TIS-associated serum lactate adversely predicts posttreatment survival of cancer patients
Although higher PDK4 expression in the tumor foci is correlated with lower survival of posttreatment in clinic settings (Fig. 2g and Supplementary Fig. 2d), whether the metabolic product lactate derived from the TME harboring stromal cells developing TIS is technically detectable and can serve as a marker for clinic purposes remains largely unclear. To address this, we acquired peripheral blood samples from PCa patients, including one cohort that experienced standard chemotherapy and the other that did not. ELISA assays of the serum from chemo-treated patients revealed that lactate levels in the treated cohort were significantly higher than that of the treatment-naïve group (Fig. 7a). The pattern was essentially reproduced by a remarkable increase of CXCL8 and SPINK1, canonical hallmarks of the SASP, in the same cohort of posttreatment patients (Fig. 7b, c). The data suggest that a circulating scale of lactate, the product of glucose metabolism via the glycolysis branch, emerges in the peripheral blood alongside with an in vivo SASP, which is intimately correlated with in situ senescence of tissues and develops after chemotherapeutic regimens, and both are systemically traceable in the serum of cancer patients. Further, it is intriguing to determine whether the blood level of lactate is correlated with that of typical SASP factors such as CXCL8 and SPINK1 in the same individual patients after clinical treatment. Subsequently analysis of ELISA data disclosed a significant and positive correlation between lactate and CXCL8, as well as between lactate and SPINK1 (Fig. 7d, e). Thus, lactate production and SASP expression is mutually linked, largely resembling the close correlation between PDK4 induction and the SASP factors as revealed by our clinical data derived from tumor samples per se (Fig. 2f).
We then expanded the study by longitudinal analysis of these factors in both primary tumor foci and peripheral blood (20 chemo-treated patients randomly selected). Surprisingly, cross-organ comparisons indicated a pronounced association between in-tissue expression and circulating level per factor, with the amounts of lactate, CXCL8 and SPINK1 apparently varying in parallel either within the primary tissue or through peripheral blood of each individual (Fig. 7f). Together, our data suggests that lactate indeed represents one of the critical TME-derived biological factors precisely imaging the development of an in vivo SASP, and can be exploited to assess the SASP magnitude in posttreatment cancer patients.
Clinical profiling subsequently uncovered a negative correlation between plasma level of lactate and posttreatment survival of PCa patients, further substantiating the pathological impact of lactate, which as a TME-derived molecule directly predicts adverse outcome once the TME is subject to irreparable damage by clinical agents (Fig. 7g). As PDK4 is subject to frequent mutation, amplification and deep deletion as disclosed by the TCGA pan-cancer atlas studies (Querying 22179 patients/22802 samples in 36 clinical studies) which document global genomics data from multiple cancer types (Cerami et al., 2012; Gao et al., 2013) (Fig. 7h), this molecule represents an important predictor of disease progression in treatment-naïve patients in clinical oncology (Li et al., 2020; Song et al., 2021). Contrasting former studies which mainly focus on the genomic alterations and pathological behaviors of cancer cells, we herein propose that routine surveillance of lactate, a major metabolic product derived from enhanced glycolysis driven by PDK4 highly expressed in stromal cells particularly those in the case of TIS, via a noninvasive avenue such as liquid biopsy, can provide a novel, practical and accurate strategy for both prognosis and prevention of advanced pathologies in clinical oncology.