Skip to main content

KLK7 expression in human tumors: a tissue microarray study on 13,447 tumors

Abstract

Background

Kallikrein-related peptidase 7 (KLK7) is a chymotrypsin-like serine protease which is essential for the desquamation of corneocytes and thus plays a pivotal role in maintaining skin homeostasis. In cancer, KLK7 overexpression was suggested to represent a route for metastasis through cleavage of cell junction and extracellular matrix proteins of cancer cells.

Methods

To comprehensively determine KLK7 protein expression in normal and neoplastic tissues, a tissue microarray containing 13,447 samples from 147 different tumor types and subtypes as well as 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry.

Results

KLK7 positivity was found in 64 of 147 tumor categories, including 17 tumor categories with at least one strongly positive case. The highest rate of KLK7 positivity was found in squamous cell carcinomas from various sites of origin (positive in 18.1%-63.8%), ovarian and endometrium cancers (4.8%-56.2%), salivary gland tumors (4.8%-13.7%), bilio-pancreatic adenocarcinomas (20.0%-40.4%), and adenocarcinomas of the upper gastrointestinal tract (3.3%-12.5%). KLK7 positivity was linked to nodal metastasis (p = 0.0005), blood vessel infiltration (p = 0.0037), and lymph vessel infiltration (p < 0.0001) in colorectal adenocarcinoma, nodal metastasis in hepatocellular carcinoma (p = 0.0382), advanced pathological tumor stage in papillary thyroid cancer (p = 0.0132), and low grade of malignancy in a cohort of 719 squamous cell carcinomas from 11 different sites of origin (p < 0.0001).

Conclusions

These data provide a comprehensive overview on KLK7 expression in normal and neoplastic human tissues. The prognostic relevance of KLK7 expression and the possible role of KLK7 as a drug target need to be further investigated.

Peer Review reports

Introduction

The human kallikrein (KLK, kallikrein-related peptidase) family refers to a group of enzymes that belong to the proteolytic family of serine proteases. To date, there are 15 known members in the human kallikrein family, designated as KLK1-KLK15 [1]. Some kallikreins have been implicated in cancer. For example, KLK3 (prostate specific antigen, PSA) is used as a blood biomarker for prostate cancer [2], and alterations of KLK4, KLK5, and KLK6 have been suggested to play a role for breast cancer aggressiveness [3]. A cancer-relevant role is also suspected for Kallikrein-related peptidase 7. KLK7 is a chymotrypsin-like serine protease with a critical role in the maintenance of skin homeostasis [4,5,6]. The KLK7 zymogen is secreted by a subset of keratinocytes into the stratum granulosum of the epidermis where it is activated by proteolytic removal of the short N-terminal [4, 7]. Activated KLK7 induces the desquamation of corneocytes from the outer layer of the keratinized squamous epithelium of the epidermis by proteolysis of corneodesmosomes [5, 8]. Dysregulated KLK7 production and secretion occurs in various skin disorders going along with dry, crusty and inflamed skin such as atopic dermatitis, Netherton syndrome, or psoriasis (summarized in [9]).

In normal tissues, KLK7 expression is largely limited to squamous epithelium [4, 5]. In cancer, however, KLK7 expression has been described to occur in a broad range of tumor entities [10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27]. Overexpression of KLK7 has been suggested to promote epithelial-mesenchymal transition (EMT) through the hydrolysis of cell membrane and extracellular matrix molecules (e.g., E-cadherin) and therefore to represent a possible mechanism for metastasis (summarized in [28]). Several studies have described high rates of KLK7 positivity in triple negative breast cancer [17], colorectal cancer [21], ovarian cancer [12,13,14], adenocarcinomas of the cervix uteri [23, 24], pancreatic cancer [10, 11] and malignant melanoma [19], but the published data on the prevalence of KLK7 immunostaining are discrepant for several tumor entities. For example, KLK7 positivity has been found in 20.0%-92.5% of adenocarcinomas of the endocervix [18, 23, 24] and from 41.0%–100% of squamous cell carcinomas of the oral cavity [20, 22, 25, 26]. Many important tumor entities such as carcinomas of the lung, stomach, esophagus, liver, bile ducts, or the endometrium have not been analyzed for KLK7 expression yet.

To better understand the prevalence and potential role of KLK7 expression in cancer, a comprehensive study analyzing large numbers of neoplastic and non-neoplastic tissues under highly standardized conditions is desirable. For this purpose, KLK7 expression was analyzed in more than 13,000 tumor tissue samples from 147 different tumor types and subtypes as well as 76 non-neoplastic tissue categories by immunohistochemistry (IHC) in a tissue microarray (TMA) format in this study.

Materials and methods

Tissue Microarrays (TMAs)

The tissue microarrays that were utilized in the current study had been described before [29,30,31]. In brief, our normal tissue TMA contains 76 different normal tissue types, and each tissue is represented by 8 samples from 8 different donors (in total: 608 samples on one slide). Our cancer TMAs were constructed from a total of 13,447 primary tumors obtained from 147 tumor types and subtypes that were distributed across 24 TMA slides. Detailed histopathological data on grade, pathological tumor stage (pT), pathological lymph node status (pN), blood vessel infiltration (V), and lymph vessel infiltration (L) were available from subsets of colon carcinomas (n = 2,351), liver carcinomas (n = 301), endometrium carcinomas (n = 259), ovarian carcinomas (n = 524), pancreas carcinomas (n = 598), papillary thyroid carcinomas (n = 382), gastric carcinomas (n = 398), as well as of 902 squamous cell carcinomas of different sites of origin. All samples were collected from the Institute of Pathology, University Hospital of Hamburg, Germany, the Institute of Pathology, Clinical Center Osnabrueck, Germany, and the Department of Pathology, Academic Hospital Fuerth, Germany. All tissues were formalin-fixed (4% buffered formalin) and subsequently embedded in paraffin. The process of TMA construction has been described earlier [32, 33]. In brief, each tissue is represented by a single 0.6 mm tissue core that was taken from a cancer containing donor block and transferred into an empty recipient paraffin block. The local ethics committee (Ethics commission Hamburg, WF-049/09) has approved the manufacturing and analysis of TMAs from archived remnants of diagnostic tissues. Patient informed consent was not required for this study. Patient data analysis without informed patient consent is covered by local laws (HmbKHG, §12). All work has been carried out in compliance with the Helsinki Declaration.

Immunohistochemistry (IHC)

Freshly cut tissue sections were used for all IHC experiments. All immunohistochemistry experiments were made manually in one day at the Institute of Pathology, University Medical Center Hamburg-Eppendorf. TMA sections of 3µ were cut, deparaffinized in xylol and rehydrated through a descending alcohol series. Immunohistochemistry was performed manually. Sections were placed for 5 min in an autoclave at 121 °C in pH 7.8 DakoTarget Retrieval Solution™ (Agilent, CA, USA) for heat induced antigen retrieval. Endogenous peroxidase was blocked in Dako Peroxidase Blocking Solution™ (Agilent, CA, USA; #52,023). Primary antibody specific for KLK7 protein (mouse monoclonal, MSVA-707M, #4860-707M, MS Validated Antibodies, Hamburg, Germany) was applied at 37 °C for 60 min at a dilution of 1:150. Specificity and cross reactivity of MSVA-707M was checked by Western blotting (Supplementary Fig. 1) and by comparison of the staining pattern with that of a second independent antibody as recommended by The International Working Group for Antibody Validation (IWGAV) [34]. For this purpose, the normal tissue TMA was also analyzed by the rabbit recombinant monoclonal KLK7 antibody EPR22594-203 (Abcam; ab254258) at a dilution of 1:7.5 and an otherwise identical protocol. As controls, skin with stratum granulosum (positive control) and colon mucosa (negative control) were included in the TMA. The EnVision detection Kit™ (Agilent, CA, USA; #K5007) was used to visualize the bound antibody. For counterstain, slides were immersed in haemalaun solution. Staining was membranous and/or cytoplasmic, which fits to the function of KLK7 as a protein that is secreted from the cytoplasm via the cell membrane to the extracellular space [28]. Accordingly, cytoplasmic and membranous staining was scored. Slide scoring was performed as described before [29,30,31]. In brief, one pathologist scored all slides (SK). In questionable cases, a second pathologist’s opinion was sought. For scoring of tumor tissues, the percentage of KLK7 positive tumor cells was estimated in each spot and the staining intensity was noted in a 4-step scale (0, 1 + , 2 + , 3 +), where 0 indicates no visble staining, 1 + ligth brown staining, 3 + dark brown staining, and 2 + staining between 1 + and 3 + . Examples for the staining intensities are given in Supplementary Fig. 2. The staining results were combined into four groups for statistical analyses: Negative staining: no visible staining, weak staining: staining intensity of 1 + in ≤ 70% or staining intensity of 2 + in ≤ 30% of tumor cells, moderate staining: staining intensity of 1 + in > 70%, staining intensity of 2 + in > 30% but in ≤ 70% or staining intensity of 3 + in ≤ 30% of tumor cells, strong staining: staining intensity of 2 + in > 70% or staining intensity of 3 + in > 30% of tumor cells.

KLK7 RNA expression data source

RNA expression z-scores of KLK7 relative to diploid samples from RNA sequencing (RNA-Seq by Expectation–Maximization (RSEM) V2, normalized from Illumina HiSeq) were downloaded from cBioportal (http://www.cbioportal.org) in July 2023. The data selection included the 32 studies labeled as “TCGA PanCancer Atlas Studies” with a total of 10,976 samples.

Western Blot

Human tissue samples of skin, kidney and liver were shock-frozen in liquid nitrogen, grinded and lysed with 5% SDS. Samples were heated for 5 min at 95 °C and the suspension was centrifuged at 10,000 g for 5 min at 8 °C. Supernatant was further used for protein quantification via Qubit protein quantification kit (Invitrogen, Carlsbad, California, USA). Protein concentration was adjusted with sample buffer (4XLDS, BioRad, Hercules, California, USA) and DTT (1:10) and the sample was heated up to 95 °C for 5 min. 30 µg per human tissue sample was used for separation on a Bis–Tris 4–20% gel. The gel was then blotted on a 0.2 µm nitrocellulose membrane. The membrane was blocked for 1 h in 5% BSA in PBST before the primary murine antibody KLK7 (MSVA-707M, MS Validated Antibodies, GmbH, Hamburg, Germany, diluted 1:1,000 in 5% BSA in PBST) was incubated on the membrane over night at 4 °C. On the next day, the membrane was incubated with the secondary antibody goat-anti-mouse-HRP (1:2,000) for 1 h at room temperature and developed with Pierce ECL Western Blotting Substrate (BioRad). Blots were imaged using a BioRad Chemidoc imager. For loading control, the membrane was incubated with a primary anti-tubulin antibody (anti-rabbit, Abcam, Cambridge, UK, ab4074) using the same protocol.

Statistics

The JMP 16 software package (SAS Institute Inc., NC, USA) was used for statistical analysis. Contingency table analysis and chi2-testing was applied to search for associations between KLK7 immunostaining and tumor phenotype. For statistical adjustment the Bonferroni correction (p = p-value/number of statistical tests) was performed, and p ≤ 0.0018 was considered as statistically significant.

Results

Technical issues

A total of 12,345 (91.8%) of 13,447 tumor samples and at least 4 normal samples per tissue category were interpretable in our TMA analysis. Non-interpretable samples demonstrated lack of unequivocal tumor cells or absence of tissue in the respective TMA spots.

KLK7 in normal tissues

KLK7 immunostaining was predominantly seen in squamous epithelia where the staining preferably occurred in a zone between the middle and the top 20% of the squamous epithelium. This area contains the granular layer in keratinizing squamous epithelium. Staining was predominantly membranous but in case of high staining intensity the cytoplasm was also involved. The extent of KLK7 staining in squamous epithelium also depended on its localization. KLK7 staining was low in the cervix uteri, higher in the esophagus and tonsil surface, and highest in the skin. In the skin, a strong staining was seen in the stratum granulosum and the keratinizing cell layers. KLK7 immunostaining was also strong in the cortex of hair follicles, the luminal area of sebaceous glands, and the central (keratinizing) zone of corpuscles of Hassall’s in the thymus. Few scattered epithelial cells or groups of cells with a moderate KLK7 positivity were occasionally seen in renal tubuli, the fallopian tube, and in salivary glands. Representative images of KLK7 positive normal tissues are shown in Fig. 1. All these findings were observed by both MSVA-707M and EPR22594-203 (Supplementary Fig. 3). An additional cytoplasmic smooth muscle staining and nuclear staining was observed in several organs for EPR22594-203 but not for MSVA-707M and was thus considered an antibody specific cross-reactivity of EPR22594-203.

Fig. 1
figure 1

KLK7 immunostaining in normal tissues. The panels show a strong membranous and cytoplasmic KLK7 staining of the granular layer of the epidermis of the skin (A) (inset shows higher magnification of the granular layer), a weak to moderate KLK7 positivity of the upper two thirds of the squamous epithelium of the esophagus (B), a strong KLK7 staining around the keratinizing zone of corpuscles of Hassall‘s of the thymus (C), and a weak to moderate KLK7 positivity in a subset of cells of squamous epithelium of tonsil crypts (D). KLK7 staining is largely absent in non-keratinizing squamous epithelium of the uterine cervix (E) and completely lacking in the mucosa of the colon (F)

KLK7 in cancer tissues

Positive KLK7 immunostaining was detectable in 1,600 (13.0%) of the 12,345 analyzable tumors, including 1,352 (11.0%) with weak, 203 (1.6%) with moderate, and 45 (0.4%) with strong immunostaining. Overall, 64 (43.5%) of 147 tumor categories showed detectable KLK7 expression with 17 (11.6%) tumor categories including at least one case with strong positivity (Table 1). Representative images of KLK7 positive tumors are shown in Fig. 2. The highest rate of KLK7 positivity was found in squamous cell carcinomas from various sites of origin (positive in 18.1%-63.8%), various categories of ovarian and endometrium cancers (4.8%-56.2%), salivary gland tumors (4.8%-13.7%), bilio-pancreatic adenocarcinomas (20.0%-40.4%), and adenocarcinomas of the upper gastrointestinal tract (3.3%-12.5%). Twenty other tumor entities showed—a usually weak—KLK7 positivity in less than 9%. A graphical representation of a ranking order of KLK7 positive and strongly positive cancers is given in Fig. 3. The relationship between KLK7 expression and clinically important histopathological and molecular tumor features in colon, liver, endometrial, ovarian, pancreas, papillary thyroid, and gastric carcinomas is shown in Table 2. Detectable KLK7 immunostaining was linked to pN + (p = 0.0005), V1 (p = 0.0037), and L1 (p < 0.0001) in colorectal adenocarcinoma, pN + in hepatocellular carcinoma (p = 0.0382), advanced pT stage in papillary thyroid cancer (p = 0.0132), and low grade of malignancy in a cohort of 719 squamous cell carcinomas from 11 different sites of origin (p < 0.0001).

Table 1 KLK7 immunostaining in human tumors
Fig. 2
figure 2

KLK7 immunostaining in cancer. The panels show a focal KLK7 immunostaining (staining intensity (int) 2 + in 10% of tumor cells) in areas of keratinization of an HPV-negative squamous cell carcinoma of the vulva (A), the oral cavity (int 1 + in 10%) (B), and of the urinary bladder (int 1 + in 20%) (C). A focal KLK7 staining is also seen in a urothelial carcinoma of the bladder with focal squamous differentiation (int 2 + in 10%) (D) while KLK7 staining is absent in a poorly differentiated squamous cell carcinoma of the esophagus lacking keratinization (int 0) (E). KLK7 positivity of variable intensity is also seen in samples from a serous high-grade carcinoma of the ovary (int 2 + in 40%) (F) as well as from an intestinal adenocarcinoma of the stomach (int 2 + in 30%) (G) and the pancreas (int 2 + in 30%) (H)

Fig. 3
figure 3

Ranking order of KLK7 immunostaining in cancers. Both the percentage of positive cases (blue dots) and the percentage of strongly positive cases (orange dots) are shown

Table 2 KLK7 immunostaining and tumor phenotype

Discussion

Our successful analysis of 12,345 tumors from 147 entities identified KLK7 expression in 64 cancer categories and enabled a ranking of tumor types according to their KLK7 positivity rate.

The most commonly KLK7 positive cancers included squamous cell carcinomas from various sites of origin, ovarian carcinomas, pancreatic adenocarcinomas, and salivary gland tumors. This is largely consistent with RNA expression data from The Cancer Genome Atlas Research Network (https://www.cancer.gov/tcga), suggesting highest rates and levels of KLK7 expression in cancers of the head and neck, the ovary and the pancreas (Fig. 4). Our data markedly expand the available data on KLK7 protein expression in cancer. Fifty of the 64 tumor categories with at least occasional KLK7 expression had not been studied by IHC before. Previous IHC studies had analyzed a total of 1,646 samples from 14 different tumor entities (summarized in Fig. 5) and often described substantially higher positivity rates than found in this study. For example, KLK7 positivity rates of ≥ 90% have been described for breast cancers [17], ovarian carcinomas [12,13,14], oral squamous cell carcinomas [20, 22, 25], adenocarcinomas of the colon [21], adenocarcinoma of the cervix [23], and different subtypes of renal cell carcinomas [27] in individual studies. Most of these discrepancies are likely to be caused by the use of different antibodies, staining protocols and definitions of thresholds to determine KLK7 positivity. Existing RNA data, along with our IHC results, do not suggest a high frequency of high KLK7 expression levels in these tumor entities [35,36,37,38]. Other reasons for discrepant findings may include variable expression in a specific molecular background. This might particularly apply for the breast cancers in our study. The general lack of detectable KLK7 expression fits well to earlier studies reporting downregulation of KLK7 mRNA in breast cancers [39, 40], but there are also studies suggesting that KLK7 can be upregulated specifically in hormone receptor negative [41] and triple negative breast cancers [17].

Fig. 4
figure 4

KLK7 in the TCGA data set and in the literature. KLK7 mRNA expression in different cancer types. Data plot generated from the cBioPortal [42, 43] database querying 10,976 samples from the “TCGA PanCancer Atlas Studies” sample set including 32 studies with KLK7 mRNA sequencing data

Fig. 5
figure 5

Comparison with previous KLK7 literature. Comparison of the percentage of samples with KLK7 expression (at least weak staining) with the corresponding data from published studies. An “X” indicates the fraction of KLK7 positive cancers in the present study, dots indicate the reported frequencies from the literature for comparison: red dots mark studies with ≤ 10 analyzed tumors, yellow dots mark studies with 11–25 analyzed tumors, and green dots mark studies with > 25 analyzed tumors. All studies are listed in the reference list

A frequent KLK7 expression in squamous cell carcinomas of various sites was expected based on the function of the protein in the desquamation of corneocytes. Desquamation of keratinized cells also occurs in cancers [44] and explains the expression of KLK7 in keratinized squamous cell carcinomas. Keratinization (or leucoplakia) is considered as a precancerous process in squamous epithelium [45,46,47] and defines a low histologic grade of malignancy in squamous cell carcinomas [48]. A significant association between KLK7 positivity and low grade could therefore be expected in these cancers. Cytokeratin 10, another protein that is specifically expressed in cornifying squamous epithelium, has also been found to be preferentially expressed in low grade squamous cell carcinomas [49]. In agreement with our observation, Leusink et al. found a link between strong KLK7 immunostaining and a favorable prognosis in a cohort of 83 squamous cell carcinomas of the oral cavity [26]. However, an association between high KLK7 protein levels and adverse pathological features or poor clinical outcome of squamous cell carcinomas has been described in two other studies on 30 and 80 cancers of the oral cavity [20, 25].

A poor prognosis of KLK7 expressing cancers could potentially be explained by in vitro and in vivo studies suggesting a relevant role of KLK7 in tumorigenesis, progression, and metastasis (summarized in [28]). The KLK7 zymogen is activated in the extracellular space to hydrolyze various extracellular matrix substrates (e.g. fibronectin 1/FN1 [50], thrombospondin 1/TSP1 [50]), cell membrane proteins (e.g. E-cadherin [11], desmoglein/DSG 1 and 2 [51]), secreted substrates (e.g. insulin growth factor binding protein/IGFBP 3 and 6 [50]), and other substrates (e.g. midkine [52], pro-matrix metalloprotease/MMP10 [50]). Studies suggested a direct or indirect effect of KLK7 on cell proliferation [19, 52], cell–cell adhesion [50, 51, 53], cell shedding [50], cell migration [50, 52, 54], cell invasion [11, 19, 52, 54], extracellular matrix organization [50, 55], EMT [11, 54], angiogenesis [28, 50], and metabolic reprogramming [28, 50] through the activation or inactivation of KLK7 substrates. For example, KLK7-mediated inactivation of E-cadherin – a hallmark of EMT [56]—and DSG1 and 2 promotes metastasis by inducing cell invasion and migration as well as reducing cell aggregation [11, 51]. KLK7-mediated inactivation of TSP1 may lead to cell proliferation, cell migration, cell invasion, and angiogenesis through the activation of the PI3K/Akt/mTOR pathway and VEGFR2 signaling [50, 57, 58]. KLK7-mediated inactivation of IGFBP3 and 6 may lead to cell proliferation through the activation of the PIK3/Akt, MEK/ERK, and Wnt/ß-Catenin pathways [50, 59] and changes in glycolysis, glycogen synthesis, and gluconeogenesis through the PI3K/Akt/mTOR pathway [50, 59, 60]. KLK7-mediated activation of proMMP10 and proMMP9 may leads to cell invasion and cell migration through the MMP-mediated hydrolysis of extracellular matrix and basement membrane components [50, 61, 62]. Furthermore, KLK7 overexpression resulted in a switch from a proliferative to an invasive phenotype in melanoma cells [19]. However, our own analyses of cancer entities with data on histomorphological features of malignancy did not provide evidence for a paramount prognostic impact of KLK7 expression on cancer malignancy. Only 7 of 28 analyses comparing KLK7 immunostaining and prognostic features resulted in significant associations. Because the number of statistical analyses was so large in our study (n = 28) that a statistical adjustment (Bonferroni correction: p ≤ 0.0018) invalidated almost all significant results, it was conspicuous that 4 of the 7 significant data involved nodal metastasis or tumor infiltration into blood or lymph vessels. These observations would indeed be consistent with a role of KLK7 associated cell dissociation in the process of metastasis.

It is an advantage of our study that only a single 0.6 mm tissue core was analyzed per cancer. The one punch per cancer approach allows for maximal standardization of the analysis because the same amount of tissue (approx. 0.28 mm2) is studied from each tumor. In contrast, the use of multiple punches per tumor almost inevitably leads to statistical bias because virtually never all samples from each tumor are interpretable. Others and we have earlier shown that one 0.6 mm core per cancer is sufficient to find clinically relevant associations between molecular markers and tumor phenotype (reviewed in [63]). For example, Torhorst et al. [64] analyzed 1–4 TMA cores each for p53, ER and PR and demonstrated that known associations between these markers and patient prognosis can be found irrespective of whether four tissue cores per tumor were analyzed separately, or a combined result was generated from the four cores.

As KLK7 is only expressed in a limited number of normal tissues and de-novo expression can occur in cancer, KLK7 may represent a suitable drug target for the treatment of cancer. Several studies have indeed demonstrated successful KLK7 inhibition by protein inhibitors and small molecule inhibitors in preclinical studies (summarized in [28]). In addition, some studies have already shown an antitumor effect of KLK7 inhibition. For example, small molecules inhibiting KLK7 have efficiently inhibited the proliferation, migration and invasion of pancreatic cancer cells [65], and cell shedding of plantar stratum corneum cells [6]. Our data suggest that ovarian, endometrial, pancreatic, and gallbladder carcinomas might represent suitable tumor entities for clinical trials once anti-KLK7 drugs will move towards testing in human patients.

Considering the large scale of our study, our assay was extensively validated by comparing our IHC findings in normal tissues with data obtained by another independent anti-KLK7 antibody and RNA data derived from three different publicly accessible databases [35,36,37,38]. To ensure an as broad as possible range of proteins to be tested for a possible cross-reactivity, 76 different normal tissues categories were included in this analysis. Validity of our assay was supported by the detection of significant KLK7 immunostaining in organs with documented KLK7 RNA expression (esophagus, salivary gland, kidney, vagina, cervix, fallopian tube, tonsil, and the skin) with the only exception of breast tissue which always stained KLK7 negative both on TMAs and on large sections. As KLK7 RNA was not detected in the FANTOM5 dataset [35, 36], it appears possible that some breast samples from other databases were contaminated by KLK7 RNAs derived from the skin covering breast tissues. KLK7 positivity in a fraction of cells in corpuscles of Hassall’s was the only IHC finding which was not matched by RNA data. This staining is conceivable, however, because of the squamous epithelial nature of corpuscles of Hassall’s. As for all other normal tissue results, KLK7 positivity of corpuscles of Hassall’s was confirmed by the use of a second independent antibody. In the thymus, the KLK7 positive cells constitute such a small fraction of the total number of cells that KLK7 RNA may not occur at detectable quantities in RNAs extracted from whole organ tissue samples. Independence of the two KLK7 antibodies used in this study is documented by cytoplasmic smooth muscle staining and nuclear staining in several organs (often predominating on nuclear membranes) which was seen by EPR22594-203 but not by MSVA-707M.

Conclusion

Our data provide a comprehensive overview on KLK7 expression in normal and neoplastic human tissues. KLK7 expression predominates in squamous cell carcinomas of different organs but is also common in ovarian and biliopancreatic adenocarcinomas as well as adenocarcinomas of the upper gastrointestinal tract. The prognostic relevance of KLK7 expression and the possible role of KLK7 as a drug target needs to be further investigated.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

IHC:

Immunohistochemistry

KLK7:

Kallikrein-related peptidase 7

pT:

Pathological tumor stage

pN:

Pathological lymph node status

TMA:

Tissue microarray

References

  1. Clements J, Hooper J, Dong Y, Harvey T. The expanded human kallikrein (KLK) gene family: genomic organisation, tissue-specific expression and potential functions. Biol Chem. 2001;382(1):5–14.

    Article  CAS  PubMed  Google Scholar 

  2. Hong SK. Kallikreins as biomarkers for prostate cancer. Biomed Res Int. 2014;2014:526341.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Avgeris M, Mavridis K, Scorilas A. Kallikrein-related peptidases in prostate, breast, and ovarian cancers: from pathobiology to clinical relevance. Biol Chem. 2012;393(5):301–17.

    Article  CAS  PubMed  Google Scholar 

  4. Yousef GM, Scorilas A, Magklara A, Soosaipillai A, Diamandis EP. The KLK7 (PRSS6) gene, encoding for the stratum corneum chymotryptic enzyme is a new member of the human kallikrein gene family - genomic characterization, mapping, tissue expression and hormonal regulation. Gene. 2000;254(1–2):119–28.

    Article  CAS  PubMed  Google Scholar 

  5. Caubet C, Jonca N, Brattsand M, Guerrin M, Bernard D, Schmidt R, Egelrud T, Simon M, Serre G. Degradation of corneodesmosome proteins by two serine proteases of the kallikrein family, SCTE/KLK5/hK5 and SCCE/KLK7/hK7. J Invest Dermatol. 2004;122(5):1235–44.

    Article  CAS  PubMed  Google Scholar 

  6. de Veer SJ, Furio L, Swedberg JE, Munro CA, Brattsand M, Clements JA, Hovnanian A, Harris JM. Selective Substrates and Inhibitors for Kallikrein-Related Peptidase 7 (KLK7) Shed Light on KLK Proteolytic Activity in the Stratum Corneum. J Invest Dermatol. 2017;137(2):430–9.

    Article  PubMed  Google Scholar 

  7. Hansson L, Strömqvist M, Bäckman A, Wallbrandt P, Carlstein A, Egelrud T. Cloning, expression, and characterization of stratum corneum chymotryptic enzyme. A skin-specific human serine proteinase. J Biol Chem. 1994;269(30):19420–6.

    Article  CAS  PubMed  Google Scholar 

  8. Levi K, Baxter J, Meldrum H, Misra M, Pashkovski E, Dauskardt RH. Effect of corneodesmosome degradation on the intercellular delamination of human stratum corneum. J Invest Dermatol. 2008;128(9):2345–7.

    Article  CAS  PubMed  Google Scholar 

  9. Kishibe M. Physiological and pathological roles of kallikrein-related peptidases in the epidermis. J Dermatol Sci. 2019;95(2):50–5.

    Article  CAS  PubMed  Google Scholar 

  10. Iakovlev V, Siegel ER, Tsao MS, Haun RS. Expression of kallikrein-related peptidase 7 predicts poor prognosis in patients with unresectable pancreatic ductal adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 2012;21(7):1135–42.

    Article  CAS  PubMed  Google Scholar 

  11. Johnson SK, Ramani VC, Hennings L, Haun RS. Kallikrein 7 enhances pancreatic cancer cell invasion by shedding E-cadherin. Cancer. 2007;109(9):1811–20.

    Article  CAS  PubMed  Google Scholar 

  12. Shigemasa K, Tanimoto H, Underwood LJ, Parmley TH, Arihiro K, Ohama K, O’Brien TJ. Expression of the protease inhibitor antileukoprotease and the serine protease stratum corneum chymotryptic enzyme (SCCE) is coordinated in ovarian tumors. Int J Gynecol Cancer. 2001;11(6):454–61.

    Article  CAS  PubMed  Google Scholar 

  13. Wang P, Magdolen V, Seidl C, Dorn J, Drecoll E, Kotzsch M, Yang F, Schmitt M, Schilling O, Rockstroh A, et al. Kallikrein-related peptidases 4, 5, 6 and 7 regulate tumour-associated factors in serous ovarian cancer. Br J Cancer. 2018;119(7):1–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dorn J, Gkazepis A, Kotzsch M, Kremer M, Propping C, Mayer K, Mengele K, Diamandis EP, Kiechle M, Magdolen V, et al. Clinical value of protein expression of kallikrein-related peptidase 7 (KLK7) in ovarian cancer. Biol Chem. 2014;395(1):95–107.

    Article  CAS  PubMed  Google Scholar 

  15. Xuan Q, Yang X, Mo L, Huang F, Pang Y, Qin M, Chen Z, He M, Wang Q, Mo ZN. Expression of the serine protease kallikrein 7 and its inhibitor antileukoprotease is decreased in prostate cancer. Arch Pathol Lab Med. 2008;132(11):1796–801.

    Article  CAS  PubMed  Google Scholar 

  16. Zhang CY, Zhu Y, Rui WB, Dai J, Shen ZJ. Expression of kallikrein-related peptidase 7 is decreased in prostate cancer. Asian J Androl. 2015;17(1):106–10.

    Article  CAS  PubMed  Google Scholar 

  17. Geng X, Babayeva L, Walch A, Aubele M, Groß E, Kiechle M, Bronger H, Dreyer T, Magdolen V, Dorn J. High levels of KLK7 protein expression are related to a favorable prognosis in triple-negative breast cancer patients. Am J Cancer Res. 2020;10(6):1785–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Santin AD, Cane’ S, Bellone S, Bignotti E, Palmieri M, De Las Casas LE, Roman JJ, Anfossi S, O’Brien T, Pecorelli S. The serine protease stratum corneum chymotryptic enzyme (kallikrein 7) is highly overexpressed in squamous cervical cancer cells. Gynecol Oncol. 2004;94(2):283–8.

    Article  CAS  PubMed  Google Scholar 

  19. Delaunay T, Deschamps L, Haddada M, Walker F, Soosaipillai A, Soualmia F, El Amri C, Diamandis EP, Brattsand M, Magdolen V, et al. Aberrant expression of kallikrein-related peptidase 7 is correlated with human melanoma aggressiveness by stimulating cell migration and invasion. Mol Oncol. 2017;11(10):1330–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kumar DV, Sivaranjani Y, Rao GV. Immunohistochemical expression of kallikrein 7 in oral squamous cell carcinoma. J Oral Maxillofac Pathol. 2020;24(3):580.

    Article  PubMed  Google Scholar 

  21. Walker F, Nicole P, Jallane A, Soosaipillai A, Mosbach V, Oikonomopoulou K, Diamandis EP, Magdolen V, Darmoul D. Kallikrein-related peptidase 7 (KLK7) is a proliferative factor that is aberrantly expressed in human colon cancer. Biol Chem. 2014;395(9):1075–86.

    Article  CAS  PubMed  Google Scholar 

  22. Pettus JR, Johnson JJ, Shi Z, Davis JW, Koblinski J, Ghosh S, Liu Y, Ravosa MJ, Frazier S, Stack MS. Multiple kallikrein (KLK 5, 7, 8, and 10) expression in squamous cell carcinoma of the oral cavity. Histol Histopathol. 2009;24(2):197–207.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Termini L, Maciag PC, Soares FA, Nonogaki S, Pereira SM, Alves VA, Longatto-Filho A, Villa LL. Analysis of human kallikrein 7 expression as a potential biomarker in cervical neoplasia. Int J Cancer. 2010;127(2):485–90.

    Article  CAS  PubMed  Google Scholar 

  24. Tian X, Shigemasa K, Hirata E, Gu L, Uebaba Y, Nagai N, O’Brien TJ, Ohama K. Expression of human kallikrein 7 (hK7/SCCE) and its inhibitor antileukoprotease (ALP/SLPI) in uterine endocervical glands and in cervical adenocarcinomas. Oncol Rep. 2004;12(5):1001–6.

    CAS  PubMed  Google Scholar 

  25. Zhao H, Dong Y, Quan J, Smith R, Lam A, Weinstein S, Clements J, Johnson NW, Gao J. Correlation of the expression of human kallikrein-related peptidases 4 and 7 with the prognosis in oral squamous cell carcinoma. Head Neck. 2011;33(4):566–72.

    Article  PubMed  Google Scholar 

  26. Leusink FK, van Diest PJ, Frank MH, Broekhuizen R, Braunius W, van Hooff SR, Willems SM, Koole R. The Co-Expression of Kallikrein 5 and Kallikrein 7 Associates with Poor Survival in Non-HPV Oral Squamous-Cell Carcinoma. Pathobiology. 2015;82(2):58–67.

    Article  CAS  PubMed  Google Scholar 

  27. Gabril M, White NM, Moussa M, Chow TF, Metias SM, Fatoohi E, Yousef GM. Immunohistochemical analysis of kallikrein-related peptidases in the normal kidney and renal tumors: potential clinical implications. Biol Chem. 2010;391(4):403–9.

    Article  CAS  PubMed  Google Scholar 

  28. Xiang F, Wang Y, Cao C, Li Q, Deng H, Zheng J, Liu X, Tan X. The Role of Kallikrein 7 in Tumorigenesis. Curr Med Chem. 2022;29(15):2617–31.

    Article  CAS  PubMed  Google Scholar 

  29. Rico SD, Mahnken M, Buscheck F, Dum D, Luebke AM, Kluth M, Hube-Magg C, Hinsch A, Hoflmayer D, Moller-Koop C, et al. MUC5AC Expression in Various Tumor Types and Nonneoplastic Tissue: A Tissue Microarray Study on 10 399 Tissue Samples. Technol Cancer Res Treat. 2021;20:15330338211043328.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Steurer S, Riemann C, Buscheck F, Luebke AM, Kluth M, Hube-Magg C, Hinsch A, Hoflmayer D, Weidemann S, Fraune C, et al. p63 expression in human tumors and normal tissues: a tissue microarray study on 10,200 tumors. Biomark Res. 2021;9(1):7.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Uhlig R, Gunther K, Broker N, Gorbokon N, Lennartz M, Dwertmann Rico S, Reiswich V, Viehweger F, Buscheck F, Kluth M, et al. Diagnostic and prognostic role of pancreatic secretory granule membrane major glycoprotein 2 (GP2) immunohistochemistry: A TMA study on 27,681 tumors. Pathol Res Pract. 2022;238:154123.

    Article  CAS  PubMed  Google Scholar 

  32. Bubendorf L, Nocito A, Moch H, Sauter G. Tissue microarray (TMA) technology: miniaturized pathology archives for high-throughput in situ studies. J Pathol. 2001;195(1):72–9.

    Article  CAS  PubMed  Google Scholar 

  33. Kononen J, Bubendorf L, Kallioniemi A, Bärlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998;4(7):844–7.

    Article  CAS  PubMed  Google Scholar 

  34. Uhlen M, Bandrowski A, Carr S, Edwards A, Ellenberg J, Lundberg E, Rimm DL, Rodriguez H, Hiltke T, Snyder M, et al. A proposal for validation of antibodies. Nat Methods. 2016;13(10):823–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lizio M, Harshbarger J, Shimoji H, Severin J, Kasukawa T, Sahin S, Abugessaisa I, Fukuda S, Hori F, Ishikawa-Kato S, et al. Gateways to the FANTOM5 promoter level mammalian expression atlas. Genome Biol. 2015;16:22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lizio M, Harshbarger J, Abugessaisa I, Noguchi S, Kondo A, Severin J, Mungall C, Arenillas D, Mathelier A, Medvedeva YA, et al. Update of the FANTOM web resource: high resolution transcriptome of diverse cell types in mammals. Nucleic Acids Res. 2017;45(D1):D737–43.

    Article  CAS  PubMed  Google Scholar 

  37. Consortium G. The Genotype-Tissue Expression (GTEx) project. Nat Genet. 2013;45(6):580–5.

    Article  Google Scholar 

  38. Karlsson M, Zhang C, Méar L, Zhong W, Digre A, Katona B, Sjöstedt E, Butler L, Odeberg J, Dusart P, et al. A single-cell type transcriptomics map of human tissues. Sci Adv. 2021;7(31):eabh2169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yousef GM, Yacoub GM, Polymeris ME, Popalis C, Soosaipillai A, Diamandis EP. Kallikrein gene downregulation in breast cancer. Br J Cancer. 2004;90(1):167–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Li X, Liu J, Wang Y, Zhang L, Ning L, Feng Y. Parallel underexpression of kallikrein 5 and kallikrein 7 mRNA in breast malignancies. Cancer Sci. 2009;100(4):601–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Talieri M, Diamandis EP, Gourgiotis D, Mathioudaki K, Scorilas A. Expression analysis of the human kallikrein 7 (KLK7) in breast tumors: a new potential biomarker for prognosis of breast carcinoma. Thromb Haemost. 2004;91(1):180–6.

    Article  CAS  PubMed  Google Scholar 

  42. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4.

    Article  PubMed  Google Scholar 

  43. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6(269):pl1.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Okegawa T, Pong RC, Li Y, Hsieh JT. The role of cell adhesion molecule in cancer progression and its application in cancer therapy. Acta Biochim Pol. 2004;51(2):445–57.

    Article  CAS  PubMed  Google Scholar 

  45. Kramer IR, Lucas RB, Pindborg JJ, Sobin LH. Definition of leukoplakia and related lesions: an aid to studies on oral precancer. Oral Surg Oral Med Oral Pathol. 1978;46(4):518–39.

    Article  CAS  PubMed  Google Scholar 

  46. van Harten AM, Brakenhoff RH. Targeted Treatment of Head and Neck (Pre) Cancer: Preclinical Target Identification and Development of Novel Therapeutic Applications. Cancers (Basel). 2021;13(11):2774.

    Article  PubMed  Google Scholar 

  47. Taggart MW, Rashid A, Ross WA, Abraham SC. Oesophageal hyperkeratosis: clinicopathological associations. Histopathology. 2013;63(4):463–73.

    Article  PubMed  Google Scholar 

  48. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, et al. The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J Thorac Oncol. 2015;10(9):1243–60.

    Article  PubMed  Google Scholar 

  49. Uhlig R, Abboud M, Gorbokon N, Lennartz M, Dwertmann Rico S, Kind S, Reiswich V, Viehweger F, Kluth M, Hube-Magg C, et al. Cytokeratin 10 (CK10) expression in cancer: A tissue microarray study on 11,021 tumors. Ann Diagn Pathol. 2022;60:152029.

    Article  PubMed  Google Scholar 

  50. Silva LM, Kryza T, Stoll T, Hoogland C, Dong Y, Stephens CR, Hastie ML, Magdolen V, Kleifeld O, Gorman JJ, et al. Integration of Two In-depth Quantitative Proteomics Approaches Determines the Kallikrein-related Peptidase 7 (KLK7) Degradome in Ovarian Cancer Cell Secretome. Mol Cell Proteomics. 2019;18(5):818–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ramani VC, Hennings L, Haun RS. Desmoglein 2 is a substrate of kallikrein 7 in pancreatic cancer. BMC Cancer. 2008;8:373.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Yu Y, Prassas I, Dimitromanolakis A, Diamandis EP. Novel Biological Substrates of Human Kallikrein 7 Identified through Degradomics. J Biol Chem. 2015;290(29):17762–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ramani VC, Haun RS. Expression of kallikrein 7 diminishes pancreatic cancer cell adhesion to vitronectin and enhances urokinase-type plasminogen activator receptor shedding. Pancreas. 2008;37(4):399–404.

    Article  CAS  PubMed  Google Scholar 

  54. Mo L, Zhang J, Shi J, Xuan Q, Yang X, Qin M, Lee C, Klocker H, Li QQ, Mo Z. Human kallikrein 7 induces epithelial-mesenchymal transition-like changes in prostate carcinoma cells: a role in prostate cancer invasion and progression. Anticancer Res. 2010;30(9):3413–20.

    CAS  PubMed  Google Scholar 

  55. Ramani VC, Haun RS. The extracellular matrix protein fibronectin is a substrate for kallikrein 7. Biochem Biophys Res Commun. 2008;369(4):1169–73.

    Article  CAS  PubMed  Google Scholar 

  56. Wong SHM, Fang CM, Chuah LH, Leong CO, Ngai SC. E-cadherin: Its dysregulation in carcinogenesis and clinical implications. Crit Rev Oncol Hematol. 2018;121:11–22.

    Article  PubMed  Google Scholar 

  57. Bazzazi H, Zhang Y, Jafarnejad M, Isenberg JS, Annex BH, Popel AS. Computer Simulation of TSP1 Inhibition of VEGF-Akt-eNOS: An Angiogenesis Triple Threat. Front Physiol. 2018;9:644.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bussolati B, Assenzio B, Deregibus MC, Camussi G. The proangiogenic phenotype of human tumor-derived endothelial cells depends on thrombospondin-1 downregulation via phosphatidylinositol 3-kinase/Akt pathway. J Mol Med (Berl). 2006;84(10):852–63.

    Article  CAS  PubMed  Google Scholar 

  59. Bach LA. What Happened to the IGF Binding Proteins? Endocrinology. 2018;159(2):570–8.

    Article  CAS  PubMed  Google Scholar 

  60. Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 2020;20(2):74–88.

    Article  CAS  PubMed  Google Scholar 

  61. Ramani VC, Kaushal GP, Haun RS. Proteolytic action of kallikrein-related peptidase 7 produces unique active matrix metalloproteinase-9 lacking the C-terminal hemopexin domains. Biochim Biophys Acta. 2011;1813(8):1525–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Davidson B, Reich R, Risberg B, Nesland JM. The biological role and regulation of matrix metalloproteinases (MMP) in cancer. Arkh Patol. 2002;64(3):47–53.

    CAS  PubMed  Google Scholar 

  63. Sauter G. Representativity of TMA studies. Methods Mol Biol. 2010;664:27–35.

    Article  CAS  PubMed  Google Scholar 

  64. Torhorst J, Bucher C, Kononen J, Haas P, Zuber M, Kochli OR, Mross F, Dieterich H, Moch H, Mihatsch M, et al. Tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am J Pathol. 2001;159(6):2249–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Du JP, Li L, Zheng J, Zhang D, Liu W, Zheng WH, Li XS, Yao RC, Wang F, Liu S, et al. Kallikrein-related peptidase 7 is a potential target for the treatment of pancreatic cancer. Oncotarget. 2018;9(16):12894–906.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to Melanie Witt, Inge Brandt, Maren Eisenberg, Laura Behm, and Sünje Seekamp for excellent technical assistance.

Funding

Open Access funding enabled and organized by Projekt DEAL.

Author information

Authors and Affiliations

Authors

Contributions

SK, FJ, LSH, CPC, MK, RS, GS contributed to conception, design, data collection, data analysis, and manuscript writing. RiS, NG, ML, SDR, SK, VR, FV, CB, FB, TSC, CF, AH, FJ, WW, AM, TK, PL, SS, EB, SM, and AHM participated in pathology data analysis, data interpretation, and collection of samples. RS, MK, CPC, CHM: data analysis. RS, GS, FJ: study supervision. All authors agree to be accountable for the content of the work.

Corresponding author

Correspondence to Ronald Simon.

Ethics declarations

Ethics approvaland consent to participate

The local ethics committee (Ethics Commission Hamburg, WF-049/09) has approved the manufacturing and analysis of TMAs from archived remnants of diagnostic tissues. Patients data analysis without informed patient consent is covered by local laws (HmbKHG, §12).

The use of archived remnants of diagnostic tissues for manufacturing of TMAs and their analysis for research purposes as well as patient data analysis has been approved by local laws (HmbKHG, §12) and by the local ethics committee (Ethics Commission Hamburg, WF-049/09). All work has been carried out in compliance with the Helsinki.

Consent for publication

Not applicable.

Competing interests

The monoclonal mouse KLK7 antibody, clone MSVA-707M was provided from MS Validated Antibodies GmbH (owned by a family member of GS). All other Authors have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kind, S., Castillo, C.P., Schlichter, R. et al. KLK7 expression in human tumors: a tissue microarray study on 13,447 tumors. BMC Cancer 24, 794 (2024). https://doi.org/10.1186/s12885-024-12552-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12552-8

Keywords