International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Oesophageal cancer is the seventh most common cancer globally, responsible for 1 in 18 cancer-related deaths and over 500,000 deaths every year (1). The two most common histological subtypes are squamous cell carcinoma and adenocarcinoma (2). Squamous cell carcinoma is the predominant type worldwide but the incidence of oesophageal adenocarcinoma (EAC) exceeds that of squamous cell carcinoma in higher-income countries (3). The 5-year survival rate of EAC is approximately 20% (4,5).
Curative treatment intents can only be performed for operable patients. For inoperable patients chemo-(radio)-therapy alone is the preferred treatment option (6). All operable EAC patients should be treated with neoadjuvant chemotherapy or chemoradiotherapy, before surgery if possible since it improves survival significantly (7,8). Therapy with a combination of cisplatin and fluorouracil the anti-HER2 monoclonal antibody trastuzumab has also been shown to be efficient in HER2-positive advanced disease (9). In addition, patients with PD-L1-positive oesophageal cancer who were treated with pembrolizumab or nivolumab had promising results. The prognosis of our patients could be improved by the introduction of neoadjuvant therapies. Today, about 20–25% of operable patients are still alive five years after primary diagnosis (10,11). However, the majority of patients die as a result of their tumour disease due to (late) relapses. The chemotherapies then available are only therapeutically effective to a very limited extent. Primarily inoperable patients or (palliative) patients with hematogenous metastases show a disastrous prognosis, only very few patients survive five years. According to national guidelines, patients who have received neoadjuvant CROSS and have no vital tumour postoperatively do not receive further adjuvant therapy. Despite improvements described above there is still a great need for therapies in the recurrent/metastatic situation. Syndecan-1 (CD138) is cell-surface heparan sulphate (12), one of four members of the syndecan family, and includes 288 amino acids which make it the second largest among (13). Syndecans play a role in adjusting cell-cell and cell-matrix interactions (14). They also play roles such as modulation of cell proliferation and invasive growth (15,16). CD138 is physiologically expressed on plasma cells and various epithelial cells (17). CD138 expression is best known to be highly specific for multiple myeloma (18). With that said, CD138 overexpression is also reported in various carcinomas such as breast, pancreatic, gallbladder, endometrial, ovarian, prostate and urinary bladder cancers (14).
Specific drugs targeting CD138 have recently been assessed in various tumours. For instance, indatuximab ravtansine, a monoclonal antibody-linked cytotoxic agent that targets CD138, is reported to be very efficient on multiple myeloma (19,20). This antibody-drug conjugate (ADC) uses the CD138 binding site on the cancer cell to hijack the cell and place the actual therapeutic agent in the tumour cell in a targeted and highly concentrated manner. The same principle of ADC is successfully used with trastuzumab deruxtecan (target protein Her2/neu) and also with sacituzumab govitecan (target protein TROP2) in breast carcinoma (21,22).
Our group recently demonstrated that sacituzumab govitecan is also effective in EAC and that the efficacy is dependent on the level of expression of TROP2 on the tumour cell (23). The same relationship was shown for CD138 in breast carcinoma. Indatuximab ravtansine led to an increased complete response in xenografts of triple-negative breast carcinomas that strongly expressed CD138 in immunohistochemistry (24,25). This makes CD138 an ideal predictive therapy-relevant biomarker. Another recently identified agent, VIS823, showed promising results in a preclinical study on multiple myeloma cell lines (26).
Almost nothing is known about the significance and expression level of CD138 in EAC. We aim to answer the following questions with this work: i) how many EACs express CD138? ii) what are the histomorphological, molecular and clinical characteristics of CD138-positive EAC? iii) is CD138 suitable as a relevant prognostic marker in EAC? iv) does neoadjuvant therapy (after CROSS or FLOT) have an impact on the expression level of CD138 in EAC?
Patients were included in this analysis if they had undergone primary surgery with curative intent for primary EAC at the Department of General Surgery Department, University of Cologne between 1996 and 2020 and if sufficient formalin-fixed paraffin-embedded material of the primary tumour was available. Demographic, histopathological and survival data were retrieved from clinical records and histopathological reports with respect to tumour characteristics including the stage of disease at the time of diagnosis according to the AJCC TNM staging system (8th edition, 2020) (27). In case of missing data on follow-up, patients were phoned to follow up in terms of the current tumour status. The study was performed according to the regulations of the Ethics Committee of the University of Cologne (approval nos. 20-1583 and 10-242).
One tissue cylinder (diameter 1.2 mm) per case was punched out from one tumour-bearing formalin-fixed, paraffin-embedded (FFPE) block using a semiautomated precision instrument. The cylinders were then transferred to an empty paraffin block. Tissue slides were stained with antibodies against CD138 (clone EP85, rabbit, 1 : 500 pretreatment with EDTA buffer, Epitomics, Burlingame, CA, USA). All immunohistochemical staining was carried out with a Leica BOND-MAX stainer (Leica Biosystems, Wetzlar, Germany) in accordance with the manufacturer's protocol. Counterstaining was done using haematoxylin and bluing reagent.
Two pathologists with special expertise in the field of EAC (DA, AQ) assessed the membranous expression of CD138 on tumour cells. A tumour cell was counted positive if ≥50% of the membrane showed CD138 expression. The percentage of tumour cells with CD138 expression in relation to all tumour cells was calculated. The categorization of our staining analyses was based on the previously published data on the subject (28). The staining intensity was determined semi-quantitatively and divided into four intensity levels (0, 1+, 2+, 3+). Four groups were then created to perform statistical analysis: tumours without expression of CD138 on their tumour cell membrane were classified as negative, tumours with a low expression intensity in ≤70% of tumour cells and moderate intensity in ≤30% of tumour cells were scored as weakly positive (1+), tumours with low staining intensity in >70% of tumour cells, moderate intensity in <30 to 70% or strong intensity in ≤30% of tumour cells were classified as moderately positive (2+) and tumours with moderate intensity in >70% of tumour cells or strong intensity in >30% were classified as strongly positive (3+). As also mentioned in the same paper, this classification corresponds to a standard categorization that has also been used by other research groups in various immunohistochemical studies (29).
Fluorescence in situ hybridization (FISH) to evaluate the ERBB2 gene amplification status was performed with a Zytolight SPEC ERBB2/CEN 17 Dual Probe Kit (Zytomed Systems GmbH, Germany) according to the manufacturer's protocol. Sample processing was performed as previously described (30).
The expression of CD138 in tumour cells was dichotomized in two ways: first, negative (0) tumours were compared to positive tumours (1–3+). In the second step, tumours with strong expression of CD138 were analysed in comparison to the other tumours (0, 1+, 2+).
The expression levels were correlated to patients' sex, age and histopathological parameters including tumour stage (pT) and lymph node status (pN0, pN+). Additionally, CD138 expression data were correlated with ERBB2-amplification status (Her2/neu) and MET status.
For the statistical comparisons, chi-square test, Fisher's exact test and one-way ANOVA were used with a Bonferroni correction for multiple comparisons. P<0.05 was considered statistically significant, P<0.1 as a statistical trend.
For survival analysis, Kaplan-Meier curves were generated and overall survival between the CD138 expression subgroups was compared using a log-rank test, or, in the case of crossing curves, a Breslow test. Additionally, multivariate Cox proportional hazard analysis was performed including prognostic factors like age, sex, tumour stage, lymph node status and the application of neoadjuvant therapy.
For all analysis and data visualization, Python v. 3.9 was used on PyCharm Community Edition 2022.2 including commonly free available packages (numpy, pandas, scipy.stats, matplotlib, pingouin, lifelines).
The total cohort included 723 patients; 632 patients were male (87.4%) and 91 were female (12.6%); 453 patients (62.7%) received neoadjuvant treatment [CROSS regime, FLOT or not further specified (NOS)]. Table I contains detailed data for the total cohort as well as patient subgroups, respectively.
When dichotomized into negative and positive tumours, the majority of EACs did not express CD138 [491 (67.9%) vs. 232 (32.1%)]. Ninety-six tumours displayed strong expression of CD138 (13.3%) (Fig. 1). These proportions were observed more or less in all treatment groups, with tumours slightly more often expressing strong levels of CD138 after neoadjuvant CROSS treatment (Tables II and III).
When CD138-positive and CD138-negative tumours were compared, CD138-positive tumours were more frequently limited to the mucosa and submucosa (pT1), while CD138-negative tumours more often showed extensive infiltration (pT2+) (P=0.008, chi-square test). Additionally, tumours expressing CD138 less often metastasized to the lymph nodes (pN0 vs. pN+) than CD138-negative tumours (P=0.014, chi-square test).
This was also observed separately for strong expression levels of CD138 compared to no, weak or moderate CD138 expression: patients with high levels of CD138 in tumour tissue had a lower tumour stage and more often no lymph node metastasis (P=0.007, P<0.001, chi-square test).
No significant association between CD138 expression and age or sex of the patients was observed (ANOVA, chi-square test; data not shown). Furthermore, there was no interdependence observed between CD138 levels and the presence or absence of neoadjuvant treatment (chi-square test, Fisher's exact test, ANOVA; data not shown).
While CD138-negative tumours only had an ERBB2 (Her2/neu) amplification in 27.9% of cases, the majority of CD138-positive tumours were Her2/neu-amplified (58.1%, P<0.001, chi-square test).
Tumour tissue with strong CD138 expression more often displayed an Her2/neu amplification than tumours with no or weaker expression (27.4% vs. 10.7%, P=0.004, chi-square test).
Tumours treated with primary surgery and CD138-positive tumours were more frequently Her2/neu-amplified as well compared to negative tumours (53.3% vs. 23.7%, P=0.014, chi-square test). The same was observed for patients who received CROSS neoadjuvant therapy (83.3% vs. 34.8%, P<0.001, chi-square test).
There was no correlation observed between MET status and CD138 expression (data not shown).
In the next step, CD138 expression was correlated with overall survival (OS). Survival data were available for 639 patients (88.4%); the minimal follow-up period included was 1 month. During the clinical follow-up, 360 patients died (56.3%). The median time of follow-up was 22.3 months (range 1–233 months).
Patients with CD138-positive tumours had a significantly longer OS than patients without CD138 expression (26.2 vs. 20.2 months, P=0.002) (Fig. 2A).
In univariate analysis, an expression of CD138 was a favourable prognostic factor (HR 0.73, 95% CI 0.59–0.92, P=0.007). When other covariates were included (age, pT, pN, ERBB2 status, MET status), CD138 expression did not remain a significant prognostic factor (HR 0.87, 95% CI 0.69–1.11, P=0.28).
When tumours with a strong CD138 expression were compared to tumours with no or weaker CD138 expression, a significant survival advantage was seen in tumours with strong CD138 expression (31.7 vs. 21.5 months, P=0.006), (Fig. 2B). In univariate analysis, strong CD138 expression was a prognostic favourable factor (HR 0.56, 95% CI 0.39–0.80, P=0.001). In multivariate Cox analysis, strong levels of CD138 were an independent favourable prognostic factor as well (HR 0.63, 95% CI 0.43–0.94, P=0.02) (Table IV).
When sub-divided by treatment, no significant advantage in OS was observed in the cohort with primary surgery or the FLOT/NOS cohort (data not shown). However, patients who received neoadjuvant CROSS therapy and had CD138-positive tumours lived significantly longer (30.75 vs. 20.75 months, P=0.008) (Fig. 2C). Patients with strong CD138 expression lived longer compared to patients with weaker intratumoural CD138 expression (33.2 vs. 23.1 months, P=0.015) (Fig. 2D). Positivity for CD138 was a favourable prognostic factor in this subgroup (HR 0.64, 95% CI 0.45–0.91, P=0.02), as well as strong expression (3+) of CD138 compared to weaker expression (HR 0.49, 95% CI 0.29–0.84, P=0.01).
In the next step, the patients were sub-divided into a cohort with mucosa/submucosa-limited tumours [(y)pT1] and patients with deeper infiltrating tumours [(y)pT2+]. For (y)pT2+ tumours, a statistical trend towards prolonged OS was observed for CD138-positive tumours and tumours with strong CD138 expression (median OS positive tumours: 21.8 months vs. 17.5 months, P=0.07; median OS tumours with strong CD138 expression: 29.1 months vs. 18.8 months, P=0.08). In multivariate analysis, strong CD138 expression was a significant favourable prognostic factor (HR 0.65, 95% CI 0.43–0.99, P=0.04).
The same effect was observed in patients without lymph node metastasis: patients with (y)pN0 tumours displayed a statistical trend for longer OS when the tumour was positive for CD138 (45.3 months vs. 38.7 months, P=0.07). When tumours had strong CD138 expression, a longer OS was observed as well (45.9 months vs. 39.9 months, P=0.08). In univariate analysis, a strong CD138 expression (3+) was a favourable prognostic factor (HR 0.52, 95% CI 0.28–0.95, P=0.03). In multivariate analysis, however, a statistical favourable trend was shown for strong expression of CD138 (HR 0.56, 95% CI 0.29–1.09, P=0.09) (Table IV).
In tumours without Her2/neu amplification, positive expression of CD138 was significantly correlated to a longer OS (23.2 months vs. 18.9 months, P=0.03). The same was observed for strong expression of CD138 compared to weaker or no expression (29.7 vs. 19.6 months, P=0.01) (Fig. 3). In multivariate analysis, strong expression of CD138 remained an independent favourable prognostic marker (HR 0.61, 95% CI 0.40–0.93, P=0.02) (Table IV).
In this study, the protein expression of CD138 and its clinical and molecular as well as prognostic significance were assessed in the largest cohort of EAC so far. We observed that CD138 expression was strong in 13.3% of the included cases. These tumours with high CD138 expression levels may be particularly amenable to targeted therapy with ADC such as indatuximab ravtansine. Whilst CD138 is best known for staining plasma cells and therefore is mainly used in the diagnostic assessment of plasma cell tumours (31), CD138 expression has been shown in up to 87% of different tumour entities, in which 71% had strong positive staining in at least one case (28). To the best of our knowledge, just one study before has evaluated the expression of CD138 in a large number of human tumours, but that included only a limited number of EACs (n=33). Based on this study, 30% of EACs (n=11) strongly expressed CD138 (28). There are several studies on CD138 expression in tumours and related prognosis: cell-surface CD138 expression has been demonstrated as favourable in mesothelioma, gastric cancer, hepatocellular carcinoma, cervical cancer and bladder cancer (32). Kusumoto et al (33) demonstrated that in ovarian cancers, epithelial CD138 expression is significantly lower in advanced disease. Additionally, in prostate adenocarcinomas, CD138 overexpression predicts early recurrence and is associated with a higher Gleason grade (34). Lendorf et al (35) found similar results in breast cancer cases, in which CD138 expression is associated with tumours of higher grade. There are several studies on CD138 expression and its prognostic significance in oesophageal squamous cell carcinomas showing CD138 expression to be higher in less invasive tumours (lower T stage) and in better differentiated tumours (36,37).
This study demonstrates for the first time that CD138 is also a favourable prognostic marker in specific patient subgroups (e.g., those with tumours resected after neoadjuvant therapy using the CROSS protocol). Since more than 90% of operable patients receive neoadjuvant therapy and the CROSS protocol is a very commonly used therapeutic regime in Europe, these findings have high clinical and therapeutical relevance. However, it must be noted that, according to the results of the Checkmate 577 study, all patients with vital tumour after surgery, who received neoadjuvant CROSS treatment, additionally receive 1 year of nivolumab; concluding prognostic statements on this specific group are not yet possible, since this therapy has only been used for a very short time (38).
Apart from CD138 being a possible prognostic indicator, therapeutical agents which specifically target CD138 could be a new treatment approach. Several of these agents have already shown promising results in some cancer types: indatuximab ravtansine (BT062) is the ADC comprising the anti-CD138 monoclonal antibody (nBT062) and the microtubule-binding cytotoxic agent maytansinoid DM4 (39); when used in multiple myeloma patients, 75% achieved a state of stable disease (40). In addition, it has been observed that in CD138-positive, triple-negative breast cell carcinoma cell lines, indatuximab ravtansine therapy is highly effective showing complete remission (41). This efficacy of indatuximab ravtansine in breast carcinoma correlated with the expression levels of CD138 on tumour cells, assessed with immunohistochemistry. This emphasizes the potential of CD138 as a predictive, therapeutically relevant biomarker for EAC as well.
The easy applicability and broad availability of a prognostic and predictive biomarker is a great advantage in practical and clinical everyday routine, as the implementation of PD-L1 and its immense therapeutical implications have demonstrated. Especially the assessment of a biomarker by immunohistochemistry, a routine procedure available in almost all pathology institutes, fulfils these requirements.
Due to the fact that CD138 is expressed in a variety of normal epithelial cells and plasma cells (28), treatment side effects could be potentially problematic; indatuximab ravtansine, however, as demonstrated by first trials, is tolerated well, with the most common adverse side effects being grade 1 or 2 (diarrhoea and fatigue) (40). Grade 3–4 adverse effects are neutropaenia, anaemia and thrombocytopaenia (42). Another newly developed agent is VIS832, a humanized IgG1-κ monoclonal antibody targeting human CD138, inducing immune cell-mediated cytotoxicity (26). The preclinical trial demonstrated a promising efficacy of VIS832 in vitro as well as in vivo (26).
Our study has some limitations. These include the retrospective nature of the analyses and the carcinomas were from a large single-tumour centre. We only studied operable patients. Future clinical studies should also determine the expression level of CD138 in endoscopic biopsies from the EAC in nonoperable or primarily hematogenously metastasized (palliative) patients. As an advantage, and this can also be understood as a prospect for future studies: We used a long-established, commercially available and well-established immunohistochemical antibody to determine CD138 on tumour cells. Analyses are thus readily reproducible. Also, clinical trials testing the efficacy of the ADC indatuximab ravtansine in EAC need to correlate therapeutic response with the expression level of CD138 on tumour cells. For this, a technique that is easy to use and also widely available in pathology institutes is helpful.
In conclusion, this is the largest and most comprehensive study on the significance of CD138 (syndecan-1) expression in EAC. We demonstrated that a significant proportion of EAC is strongly CD138-positive (13.3%). CD138 is already utilized by ADCs such as indatuximab ravtansine, whose effectiveness depends on the extent of CD138 on tumour cells. This makes CD138 an ideal predictive, therapeutically relevant biomarker. Future clinical trials now need to show how effective the corresponding ADCs are in CD138-positive EACs.
Not applicable.
Funding: No funding was received.
The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.
DA, AS, RB and AQ made substantial contributions to conception and design. DA, AS, CB and FG were responsible for analysis and interpretation of data. DA, AS, TZ and AQ wrote the main manuscript. CB, WS and TZ were responsible for the data collection. TZ, WS and RB have reviewed the text. All authors were involved in drafting the manuscript or revising it critically for important intellectual content. All authors read and approved the final manuscript. AQ, DA and AS confirm the authenticity of all the raw data.
The objective of the project is primarily in the field of diagnostics and quality assurance; approval was obtained from the University of Cologne Ethics Committee (approval nos. 20-1583 and 10-242). All authors confirm that methods used were carried out in accordance with relevant guidelines and regulations. The experimental protocols were approved by the licensing committees. We confirm that written informed consent was obtained from all subjects and/or their legal guardians.
Not applicable.
The authors declare that they have no competing interests.
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pennathur A, Gibson MK, Jobe BA and Luketich JD: Oesophageal carcinoma. Lancet. 381:400–412. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Lepage C, Rachet B, Jooste V, Faivre J and Coleman MP: Continuing rapid increase in esophageal adenocarcinoma in England and Wales. Am J Gastroenterol. 103:2694–2699. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Tustumi F, Kimura CM, Takeda FR, Uema RH, Salum RA, Ribeiro-Junior U and Cecconello I: Prognostic factors and survival analysis in esophageal carcinoma. Arq Bras Cir Dig. 29:138–141. 2016.(In English, Portuguese). View Article : Google Scholar : PubMed/NCBI | |
|
He H, Chen N, Hou Y, Wang Z, Zhang Y, Zhang G and Fu J: Trends in the incidence and survival of patients with esophageal cancer: A SEER database analysis. Thorac Cancer. 11:1121–1128. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lagergren J and Lagergren P: Oesophageal cancer. BMJ. 341:c62802010. View Article : Google Scholar : PubMed/NCBI | |
|
Surgical resection with or without preoperative chemotherapy in oesophageal cancer, . A randomised controlled trial. Lancet. 359:1727–1733. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Gebski V, Burmeister B, Smithers BM, Foo K, Zalcberg J and Simes J; Australasian Gastro-Intestinal Trials Group, : Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: A meta-analysis. Lancet Oncol. 8:226–234. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, et al: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 376:687–697. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Doi T, Piha–Paul SA, Jalal SI, Mai–Dang H, Saraf S, Csiki MK and Bennouna J: Updated results for the advanced esophageal carcinoma cohort of the phase 1b KEYNOTE-028 study of pembrolizumab. J Clin Oncol. 34 (Suppl 15):S4046. 2016. View Article : Google Scholar | |
|
Piro G, Carbone C, Santoro R, Tortora G and Melisi D: Predictive biomarkers for the treatment of resectable esophageal and esophago-gastric junction adenocarcinoma: From hypothesis generation to clinical validation. Expert Rev Mol Diagn. 18:357–370. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bernfield M, Götte M, Park PW, Reizes O, Fitzgerald ML, Lincecum J and Zako M: Functions of cell surface heparan sulfate proteoglycans. Annu Rev Biochem. 68:729–777. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Czarnowski D: Syndecans in cancer: A review of function, expression, prognostic value, and therapeutic significance. Cancer Treat Res Commun. 27:1003122021. View Article : Google Scholar : PubMed/NCBI | |
|
Palaiologou M, Delladetsima I and Tiniakos D: CD138 (syndecan-1) expression in health and disease. Histol Histopathol. 29:177–189. 2014.PubMed/NCBI | |
|
Nikolova V, Koo CY, Ibrahim SA, Wang Z, Spillmann D, Dreier R, Kelsch R, Fischgräbe J, Smollich M, Rossi LH, et al: Differential roles for membrane-bound and soluble syndecan-1 (CD138) in breast cancer progression. Carcinogenesis. 30:397–407. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Hassan H, Greve B, Pavao MS, Kiesel L, Ibrahim SA and Götte M: Syndecan-1 modulates β-integrin-dependent and interleukin-6-dependent functions in breast cancer cell adhesion, migration, and resistance to irradiation. FEBS J. 280:2216–2227. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fears CY and Woods A: The role of syndecans in disease and wound healing. Matrix Biol. 25:443–456. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Dhodapkar MV, Abe E, Theus A, Lacy M, Langford JK, Barlogie B and Sanderson RD: Syndecan-1 is a multifunctional regulator of myeloma pathobiology: Control of tumor cell survival, growth, and bone cell differentiation. Blood. 91:2679–2688. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Schönfeld K, Zuber C, Pinkas J, Häder T, Bernöster K and Uherek C: Indatuximab ravtansine (BT062) combination treatment in multiple myeloma: Pre-clinical studies. J Hematol Oncol. 10:132017. View Article : Google Scholar : PubMed/NCBI | |
|
Kelly KR, Chanan–Khan A, Heffner LT, Somlo G, Siegel DS, Zimmerman T, Karnad A, Munshi NC, Jagannath S, Greenberg AL, et al: Indatuximab ravtansine (BT062) in combination with lenalidomide and low-dose dexamethasone in patients with relapsed and/or refractory multiple myeloma: Clinical activity in patients already exposed to lenalidomide and bortezomib. Blood. 124:47362014. View Article : Google Scholar | |
|
Koster KL, Huober J and Joerger M: New antibody-drug conjugates (ADCs) in breast cancer-an overview of ADCs recently approved and in later stages of development. Explor Target Antitumor Ther. 3:27–36. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Rugo HS, Tolaney SM, Loirat D, Punie K, Bardia A, Hurvitz SA, O'Shaughnessy J, Cortés J, Diéras V, Carey LA, et al: Safety analyses from the phase 3 ASCENT trial of sacituzumab govitecan in metastatic triple-negative breast cancer. NPJ Breast Cancer. 8:982022. View Article : Google Scholar : PubMed/NCBI | |
|
Hoppe S, Meder L, Gebauer F, Ullrich RT, Zander T, Hillmer AM, Buettner R, Plum P, Puppe J, Malter W and Quaas A: Trophoblast cell surface Antigen 2 (TROP2) as a predictive bio-marker for the therapeutic efficacy of sacituzumab govitecan in adenocarcinoma of the esophagus. Cancers (Basel). 14:47892022. View Article : Google Scholar : PubMed/NCBI | |
|
Ibrahim SA, Hassan H, Vilardo L, Kumar SK, Kumar AV, Kelsch R, Schneider C, Kiesel L, Eich HT, Zucchi I, et al: Syndecan-1 (CD138) modulates triple-negative breast cancer stem cell properties via regulation of LRP-6 and IL-6-mediated STAT3 signaling. PLoS One. 8:e857372013. View Article : Google Scholar : PubMed/NCBI | |
|
Schönfeld K, Herbener P, Zuber C, Häder T, Bernöster K, Uherek C and Schüttrumpf J: Activity of indatuximab ravtansine against triple-negative breast cancer in preclinical tumor models. Pharm Res. 35:1182018. View Article : Google Scholar : PubMed/NCBI | |
|
Yu T, Chaganty B, Lin L, Xing L, Ramakrishnan B, Wen K, Hsieh PA, Wollacott A, Viswanathan K, Adari H, et al: VIS832, a novel CD138-targeting monoclonal antibody, potently induces killing of human multiple myeloma and further synergizes with IMiDs or bortezomib in vitro and in vivo. Blood Cancer J. 10:1102020. View Article : Google Scholar : PubMed/NCBI | |
|
Brierley JD, Gospodarowicz MK and Wittekind C: TNM classification of malignant tumours. John Wiley & Sons; 2017 | |
|
Kind S, Merenkow C, Büscheck F, Möller K, Dum D, Chirico V, Luebke AM, Höflmayer D, Hinsch A, Jacobsen F, et al: Prevalence of Syndecan-1 (CD138) expression in different kinds of human tumors and normal tissues. Dis Markers. 2019:49283152019. View Article : Google Scholar : PubMed/NCBI | |
|
Simon R, Mirlacher M and Sauter G: Immunohistochemical analysis of tissue microarrays. Methods Mol Biol. 664:113–126. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Plum PS, Gebauer F, Krämer M, Alakus H, Berlth F, Chon SH, Schiffmann L, Zander T, Büttner R, Hölscher AH, et al: HER2/neu (ERBB2) expression and gene amplification correlates with better survival in esophageal adenocarcinoma. BMC Cancer. 19:382019. View Article : Google Scholar : PubMed/NCBI | |
|
Chilosi M, Adami F, Lestani M, Montagna L, Cimarosto L, Semenzato G, Pizzolo G and Menestrina F: CD138/syndecan-1: A useful immunohistochemical marker of normal and neoplastic plasma cells on routine trephine bone marrow biopsies. Mod Pathol. 12:1101–1106. 1999.PubMed/NCBI | |
|
Szatmári T, Ötvös R, Hjerpe A and Dobra K: Syndecan-1 in cancer: Implications for cell signaling, differentiation, and prognostication. Dis Markers. 2015:7960522015. View Article : Google Scholar : PubMed/NCBI | |
|
Kusumoto T, Kodama J, Seki N, Nakamura K, Hongo A and Hiramatsu Y: Clinical significance of syndecan-1 and versican expression in human epithelial ovarian cancer. Oncol Rep. 23:917–925. 2010.PubMed/NCBI | |
|
Zellweger T, Ninck C, Mirlacher M, Annefeld M, Glass AG, Gasser TC, Mihatsch MJ, Gelmann EP and Bubendorf L: Tissue microarray analysis reveals prognostic significance of syndecan-1 expression in prostate cancer. Prostate. 55:20–29. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Lendorf ME, Manon-Jensen T, Kronqvist P, Multhaupt HA and Couchman JR: Syndecan-1 and syndecan-4 are independent indicators in breast carcinoma. J Histochem Cytochem. 59:615–629. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Mikami S, Ohashi K, Usui Y, Nemoto T, Katsube K, Yanagishita M, Nakajima M, Nakamura K and Koike M: Loss of syndecan-1 and increased expression of heparanase in invasive esophageal carcinomas. Jpn J Cancer Res. 92:1062–1073. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Szumilo J, Burdan F, Zinkiewicz K, Dudka J, Klepacz R, Dabrowski A and Korobowicz E: Expression of syndecan-1 and cathepsins D and K in advanced esophageal squamous cell carcinoma. Folia Histochem Cytobiol. 47:571–578. 2009.PubMed/NCBI | |
|
Horiba MN, Casak SJ, Mishra-Kalyani PS, Roy P, Beaver JA, Pazdur R, Kluetz PG, Lemery SJ and Fashoyin-Aje LA: FDA approval summary: Nivolumab for the adjuvant treatment of adults with completely resected esophageal/gastroesophageal junction cancer and residual pathologic disease. Clin Cancer Res. 28:5244–5248. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ikeda H, Hideshima T, Fulciniti M, Lutz RJ, Yasui H, Okawa Y, Kiziltepe T, Vallet S, Pozzi S, Santo L, et al: The monoclonal antibody nBT062 conjugated to cytotoxic Maytansinoids has selective cytotoxicity against CD138-positive multiple myeloma cells in vitro and in vivo. Clin Cancer Res. 15:4028–4037. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Jagannath S, Heffner LT Jr, Ailawadhi S, Munshi NC, Zimmerman TM, Rosenblatt J, Lonial S, Chanan-Khan A, Ruehle M, Rharbaoui F, et al: Indatuximab ravtansine (BT062) monotherapy in patients with relapsed and/or refractory multiple myeloma. Clin Lymphoma Myeloma Leuk. 19:372–380. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Schönfeld K, Herbener P, Zuber C, Häder T, Bernöster K, Uherek C and Schüttrumpf J: Activity of indatuximab ravtansine against triple-negative breast cancer in preclinical tumor models. Pharm Res. 35:1182018. View Article : Google Scholar : PubMed/NCBI | |
|
Kelly KR, Ailawadhi S, Siegel DS, Heffner LT, Somlo G, Jagannath S, Zimmerman TM, Munshi NC, Madan S, Chanan-Khan A, et al: Indatuximab ravtansine plus dexamethasone with lenalidomide or pomalidomide in relapsed or refractory multiple myeloma: A multicentre, phase 1/2a study. Lancet Haematol. 8:e794–e807. 2021. View Article : Google Scholar : PubMed/NCBI |