Acessibilidade / Reportar erro

Density of high endothelial venules and PDL-1 expression: relationship with tumor-infiltrating lymphocytes in primary cutaneous melanomas

Abstract

Studies have highlighted melanoma immunogenicity, and the prognostic importance of tumor infiltrating lymphocytes (TILs) and mechanisms of tumor immune evasion, such as hyperexpression of programmed cell death ligand 1 (PDL-1). High endothelial venules (HEV) are specialized blood vessels that can facilitate the lymphocytes migration to the tumor. Here we evaluate the association of HEV density and PDL-1 expression in primary cutaneous melanomas with the presence and degree of TILs and with other clinicopathological variables (age, sex, tumor location, melanoma histological type, Breslow thickness, ulceration, regression signs, mitotic index). HEV density and PDL-1 expression were assessed immunohistochemically in 78 melanoma cases, using a specific antibody, and were detected in 59% and 76% of these, respectively. Positive associations were identified between HEV density and PDL-1 expression with the presence and degree of lymphocytic infiltration, melanoma histological type and ulceration presence. No correlation was found between HEV density and PDL-1 expression. Our findings confirm the HEV role in the recruitment and facilitation of lymphocyte transport in cutaneous melanomas, where HEV density is strongly associated with the degree of TILs. Additionally, PDL-1 hyperexpression suggests a possible mechanism of tumor immune evasion, which may lead to inactivation and reduction of the tumor lymphocytes number.

Key words
MECA-79; Melanoma; PDL-1M; tumor infiltrating lymphocytes; tumor microenvironment

INTRODUCTION

Cutaneous melanomas are aggressive neoplasms that derive from melanocytes and have a high metastatic potential. Mortality rates are high and have increased worldwide in recent decades (Taylor et al. 2007TAYLOR RC, PATEL A, PANAGEAS KS, BUSAM KJ & BRADY MS. 2007. Tumor-Infiltrating Lymphocytes Predict Sentinel Lymph Node Positivity in Patients with Cutaneous Melanoma. J Clin Oncol 25: 869-875., Fortes et al. 2015FORTES C, MASTROENI S, MANNOORANPARAMPIL TJ, PASSARELLI F, ZAPPALA A & ANNESSI G. 2015. Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res 25: 306-311., Keun Park et al. 2017KEUN PARK C, KYUM KIM S, KEUN PARK C & KYUM KIM S. 2017. Clinicopathological significance of intratumoral and peritumoral lymphocytes and lymphocyte score based on the histologic subtypes of cutaneous melanoma. Oncotarget 8: 14759-14769.).

Several studies have highlighted the immunogenic capacity of melanoma and the importance of tumor infiltrating lymphocytes (TILs) as a manifestation of host immune response (Taylor et al. 2007TAYLOR RC, PATEL A, PANAGEAS KS, BUSAM KJ & BRADY MS. 2007. Tumor-Infiltrating Lymphocytes Predict Sentinel Lymph Node Positivity in Patients with Cutaneous Melanoma. J Clin Oncol 25: 869-875., Azimi et al. 2012AZIMI F, SCOLYER RA, RUMCHEVA P, MONCRIEFF M, MURALI R & MCCARTHY SW. 2012. Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol 30: 2678-2683., Thomas et al. 2013THOMAS NE, BUSAM KJ, FROM L, KRICKER A, ARMSTRONG BK & ANTON-CULVER H. 2013. Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J Clin Oncol 31: 4252-4259., Duprat et al. 2016DUPRAT JP, BRECHTBÜLH ER, COSTA DE SÁ B, ENOKIHARA M, FREGNANI JH & LANDMAN G. 2016. Absence of Tumor-Infiltrating Lymphocyte Is a Reproducible Predictive Factor for Sentinel Lymph Node Metastasis: A Multicenter Database Study by the Brazilian Melanoma Group. PLoS ONE 11 : e0148160., Maibach et al. 2020MAIBACH F, SADOZAI H, SEYED JAFARI SM, HUNGER RE & SCHENK M. 2020. Tumor-infiltrating lymphocytes and their prognostic value in cutaneous melanoma. Frontiers Immunol 11: 2105.). There is evidence that the presence and degree of TILs, in both primary and metastatic cutaneous melanomas, has a favorable impact on melanoma overall survival, with a lower chance of developing regional and distant lymph node metastases, and is a strong marker of immune response (Clemente et al. 1996CLEMENTE CG, MIHM MC, BUFALINO R, ZURRIDA S, COLLINI P & CASCINELLI N. 1996. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77: 1303-1310., Taylor et al. 2007TAYLOR RC, PATEL A, PANAGEAS KS, BUSAM KJ & BRADY MS. 2007. Tumor-Infiltrating Lymphocytes Predict Sentinel Lymph Node Positivity in Patients with Cutaneous Melanoma. J Clin Oncol 25: 869-875., Azimi et al. 2012AZIMI F, SCOLYER RA, RUMCHEVA P, MONCRIEFF M, MURALI R & MCCARTHY SW. 2012. Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol 30: 2678-2683., Thomas et al. 2013THOMAS NE, BUSAM KJ, FROM L, KRICKER A, ARMSTRONG BK & ANTON-CULVER H. 2013. Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J Clin Oncol 31: 4252-4259., Fortes et al. 2015FORTES C, MASTROENI S, MANNOORANPARAMPIL TJ, PASSARELLI F, ZAPPALA A & ANNESSI G. 2015. Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res 25: 306-311., Duprat et al. 2016DUPRAT JP, BRECHTBÜLH ER, COSTA DE SÁ B, ENOKIHARA M, FREGNANI JH & LANDMAN G. 2016. Absence of Tumor-Infiltrating Lymphocyte Is a Reproducible Predictive Factor for Sentinel Lymph Node Metastasis: A Multicenter Database Study by the Brazilian Melanoma Group. PLoS ONE 11 : e0148160., Keun Park et al. 2017KEUN PARK C, KYUM KIM S, KEUN PARK C & KYUM KIM S. 2017. Clinicopathological significance of intratumoral and peritumoral lymphocytes and lymphocyte score based on the histologic subtypes of cutaneous melanoma. Oncotarget 8: 14759-14769.). In this context, the possible role of high endothelial venules (HEV) in melanomas and other solid tumors has been highlighted (Hayasaka et al. 2010HAYASAKA H, TANIGUCHI K, FUKAI S & MIYASAKA M. 2010. Neogenesis and development of the high endothelial venules that mediate lymphocyte trafficking. Cancer Sci 101: 2302-2308., Martinet et al. 2011MARTINET L, GARRIDO I, FILLERON T, GUELLEC S LE, BELLARD E & FOURNIE JJ. 2011. Human solid tumors contain high endothelial venules: Association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res 71: 5678-5687., 2012a, Avram et al. 2013AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861., Gallimore et al. 2013GALLIMORE A, GODKIN A & AGER A. 2013. High Endothelial Venules, Help or hindrance in the quest for antitumor immunity? Oncoimmunology 2(5): e24272., Perivoliotis et al. 2017PERIVOLIOTIS K, NTELLAS P, DADOULI K, KOUTOUKOGLOU P, IOANNOU M & TEPETES K. 2017. Microvessel Density in Patients with Cutaneous Melanoma: An Up-to-Date Systematic Review and Meta-Analysis. J Skin Cancer 2017., Asrir et al. 2022ASRIR A ET AL. 2022. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell 3: 318-334.). Investigations into lymphocyte recirculation led to the discovery of L-selectin as a receptor on lymphocytes, involved in their interaction with HEV in lymph nodes. Further studies have shown that L-selectin mediates the interaction and circulation of lymphocytes through HEVs, as the first step in the adhesion signaling cascade that culminates in the recruitment of lymphocytes to lymph nodes (Hemmerich et al. 1994HEMMERICH S, BUTCHER EC & ROSEN SD. 1994. Sulfation-dependent recognition of high endothelial venules (HEV)-ligands by L-selectin and MECA 79, and adhesion-blocking monoclonal antibody. J Exp Med 180: 2219-2226., Drayton et al. 2003DRAYTON DL, YING X, LEE J, LESSLAUER W & RUDDLE NH. 2003. Ectopic LTO: Directs Lymphoid Organ Neogenesis with Concomitant Expression of Peripheral Node Addressin and a HEV-restricted Sulfotransferase. J Experiment Med 197(9): 1153-1163., Uchimura & Rosen 2006UCHIMURA K & ROSEN SD. 2006. Sulfated L-selectin ligands as a therapeutic target in chronic inflammation. Trends Immunol 27: 559-565., Weinstein & Storkus 2016WEINSTEIN AM & STORKUS WJ. 2016. Biosynthesis and functional significance of peripheral node addressin in cancer-associated TLO. Front Immunol 7: 1-8.). The presence and density of these venules can be assessed by the expression of the specific marker MECA-79 (Michie et al. 1993MICHIE SA, STREETER PR, BOLT PA, BUTCHER EC & PICKER LJ. 1993. The Human Peripheral Lymph Node Vascular Addressin An Inducible Endothelial Antigen Involved in Lymphocyte Homing. Am J Pathol 143: 1688-1698., Miyasaka & Tanaka 2004MIYASAKA M & TANAKA T. 2004. Lymphocyte trafficking across high endothelial venules: dogmas and enigmas. Nat Rev Immunol 1: 360., Middleton et al. 2005MIDDLETON J, AMERICH L, GAYON R, JULIEN D, MANSAT M & MANSAT P. 2005. A comparative study of endothelial cell markers expressed in chronically inflamed human tissues: MECA-79, Duffy antigen receptor for chemokines, von Willebrand factor, CD31, CD34, CD105 and CD146. J Pathol 206: 260-268., Sinha et al. 2006SINHA RK, YANG G, ALEXANDER C & MAGE RG. 2006. De novo expression of MECA-79 glycoprotein-determinant on developing B lymphocytes in gut-associated lymphoid tissues. Immunology 119: 461-469., Martinet et al. 2012bMARTINET L, LE GUELLEC S, FILLERON T, LAMANT L, MEYER N & ROCHAIX P. 2012a. High endothelial venules (HEVs) in human melanoma lesions: Major gateways for tumor-infiltrating lymphocytes. Oncoimmunology 1: 829-839., Avram et al. 2013AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861.). The discovery of MECA-79, a monoclonal antibody (mAb) that reacts with the family of sialomucins collectively known as Peripheral Node Addressin (PNAd), gave rise to a powerful tool for the study of HEVs that express ligands for L-selectin (Michie et al. 1993MICHIE SA, STREETER PR, BOLT PA, BUTCHER EC & PICKER LJ. 1993. The Human Peripheral Lymph Node Vascular Addressin An Inducible Endothelial Antigen Involved in Lymphocyte Homing. Am J Pathol 143: 1688-1698., Middleton et al. 2005MIDDLETON J, AMERICH L, GAYON R, JULIEN D, MANSAT M & MANSAT P. 2005. A comparative study of endothelial cell markers expressed in chronically inflamed human tissues: MECA-79, Duffy antigen receptor for chemokines, von Willebrand factor, CD31, CD34, CD105 and CD146. J Pathol 206: 260-268., Martinet et al. 2012aMARTINET L, GARRIDO I & GIRARD JP. 2012b. Tumor high endothelial venules (HEVs) predict lymphocyte infiltration and favorable prognosis in breast cancer. Oncoimmunology 1: 789-790.).

Given the diversity of tumor microenvironments, several mechanisms promote tumor immune evasion, resulting in tumor progression (Sapoznik et al. 2012SAPOZNIK S, HAMMER O, ORTENBERG R, BESSER MJ, BEN-MOSHE T & SCHACHTER J. 2012. Novel anti-melanoma immunotherapies: Disarming tumor escape mechanisms. Clin Dev Immunol 2012: 818214., Quail & Joyce 2013QUAIL DF & JOYCE JA. 2013. Microenvironmental regulation of tumor progression and metastasis. Nat Med 19: 1423-1437., Taube et al. 2013TAUBE JM, ANDERS RA, YOUNG GD, XU H, SHARMA R & MCMILLER TL. 2013. Mechanism of Immune Escape. Sci Transl Med 4: 1-22.). One such mechanism is the overexpression of programmed cell death ligand 1 (PDL-1). The binding of PDL-1 to its programmed cell death protein 1 (PD-1) receptor in immune cells inactivates the TILs in the tumor microenvironment. This suggests that PDL-1 expression in tumor cells, evaluated by immunohistochemistry (IHC), has an adverse prognostic value, and reinforces its association with TILs in a variety of solid tumors, including melanoma (Hino et al. 2010HINO R, KABASHIMA K, KATO Y, YAGI H, NAKAMURA M & HONJO T. 2010. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 116: 1757-1766., Kashani-Sabet 2010KASHANI-SABET M. 2010. Tumor progression by immune evasion in melanoma: Role of the programmed cell death-1/programmed cell death-1 ligand 1 interaction. Cancer 116: 1623-1625., Leite et al. 2015LEITE KR, REIS ST, JUNIOR JP, ZERATI M, GOMES DDE O & CAMARA-LOPES LH. 2015. PD-L1 expression in renal cell carcinoma clear cell type is related to unfavorable prognosis. Diagn Pathol 10: 189., Massi et al. 2014MASSI D, BRUSA D, MERELLI B, CIANO M, AUDRITO V & SERRA S. 2014. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann Oncol 25: 2433-2442., Muenst et al. 2014MUENST S, SCHAERLI AR, GAO F, DUSTER S, TRELLA E & DROESER RA. 2014. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 146: 15-24., Schalper 2014SCHALPER KA. 2014. PD-L1 expression and tumor-infiltrating lymphocytes: Revisiting the antitumor immune response potential in breast cancer. Oncoimmunology 3: e29288., Velcheti et al. 2014VELCHETI V, SCHALPER KA, CARVAJAL DE, ANAGNOSTOU VK, SYRIGOS KN & SZNOL M. 2014. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Investig 94: 107-116., Jin et al. 2015JIN Y, ZAHO J, SHI X & YU X. 2015. Prognostic value of programed death ligand 1 in patients with solid tumors: A meta-analysis. J Cancer Res Ther 11: C38-C43., Wimberly et al. 2015WIMBERLY H, BROWN JR, SCHALPER K, HAACK H, SILVER MR & NIXON C. 2015. PD-L1 Expression Correlates with Tumor-Infiltrating Lymphocytes and Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancer Immunol Res 3: 326-332., Wu et al. 2015WU P, WU D, LI L, CHAI Y & HUANG J. 2015. PD-L1 and survival in solid tumors: A meta-analysis. PLoS ONE 10: 1-15., Xu et al. 2015XU F, XU L, WANG Q, AN G, FENG G & LIU F. 2015. Clinicopathological and prognostic value of programmed death ligand-1 (PD-L1) in renal cell carcinoma: a meta-analysis. Int J Clin Exp Med 8: 14595-14603., Beckers et al. 2016BECKERS RK, SELINGER CI, VILAIN R, MADORE J, WILMOTT JS & HARVEY K. 2016. Programmed death ligand 1 expression in triple-negative breast cancer is associated with tumour-infiltrating lymphocytes and improved outcome. Histopathology 69: 25-34., Zeng et al. 2016ZENG J, ZHANG XK, CHEN HD, ZHONG ZH, WU QL & LIN SX. 2016. Expression of programmed cell death-ligand 1 and its correlation with clinical outcomes in gliomas. Oncotarget 7(8): 8944., Juneja et al. 2017JUNEJA VR, MCGUIRE KA, MANGUSO RT, LAFLEUR MW, COLLINS N & HAINING WN. 2017. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med 214: 895-904.). In this context, immunotherapy studies have shown that blocking the PD-1/PDL-1 axis can reactivate the antitumor immune response, with clinical benefits (Hasan et al. 2011HASAN A, GHEBEH H, LEHE C, AHMAD R & DERMIME S. 2011. Therapeutic targeting of B7-H1 in breast cancer. Expert Opin Ther Targets 15: 1211-1225., Sapoznik et al. 2012SAPOZNIK S, HAMMER O, ORTENBERG R, BESSER MJ, BEN-MOSHE T & SCHACHTER J. 2012. Novel anti-melanoma immunotherapies: Disarming tumor escape mechanisms. Clin Dev Immunol 2012: 818214., Sanlorenzo et al. 2014SANLORENZO M, VUJIC I, POSCH C, DAJEE A, YEN A & KIM S. 2014. Melanoma immunotherapy. Cancer Biol Ther 15: 665-674., Ascierto & Marincola 2015ASCIERTO PA & MARINCOLA FM. 2015. EBioMedicine 2015: The Year of Anti-PD-1 / PD-L1s Against Melanoma and Beyond. EBIOM 2: 92-93., Philips & Atkins 2015PHILIPS GK & ATKINS M. 2015. Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Int Immunol 27: 39-46., Chen et al. 2016CHEN R, PENG PC, WEN B, LI FY, XIE S & CHEN G. 2016. Anti–Programmed Cell Death (PD)-1 Immunotherapy for Malignant Tumor: A Systematic Review and Meta-Analysis. Transl Oncol 9: 32-40., Daud et al. 2016DAUD AI, WOLCHOK JD, ROBERT C, HWU WJ, WEBER JS & RIBAS A. 2016. Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol 34: 4102-4109., Carbognin et al. 2015CARBOGNIN L, PILOTTO S, MILELLA M, VACCARO V & BRUNELLI M. 2015. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourin Cancers. PLoS ONE 10(6): e0130142.).

The aim of this study was to determine not only HEV density and PDL-1 expression in primary cutaneous melanomas, but also the association of these markers with the presence and degree of TILs and with other clinicopathological variables such as age, sex, tumor location, melanoma histological type, Breslow thickness, ulceration, regression signs and mitotic index. To the best of our knowledge, no studies to date have focused on establishing the association of HEV density with PDL-1 expression in primary cutaneous melanomas.

MATERIALS AND METHODS

Case Selection

A retrospective study was conducted to analyze 78 samples from 76 patients with primary cutaneous melanoma, stored in a pathology laboratory in southern Brazil. This study was approved by the Ethics Committee for Research with Human Beings of the University of Passo Fundo (CEP: 78759317.9.0000.5342). The cases were selected from specimens taken only from the complete resection of lesions, which consisted of superficial spreading melanoma (SSM), nodular melanoma (NM), lentigo maligna melanoma (LMM) or acral lentiginous melanoma (ALM). Samples from incisional biopsy, in situ melanomas, or melanomas not classified in the four above described histological types, or samples contained within little paraffin block material, were excluded from the study. The clinical characteristics analyzed here included the patient’s age and sex and anatomical site of the lesion.

Histopathological Analysis

Melanoma was diagnosed by analyzing tumor sections stained by hematoxylin and eosin (H&E), under an optical microscope, in samples fixed in 10% buffered formalin and embedded in paraffin. Histopathological characteristics such as: histological type (SSM, NM, LMM and ALM), Clark level, Breslow thickness, presence and degree of lymphocytic infiltration, ulceration, mitosis and signs of tumor regression were analyzed. Breslow thickness was assessed according to the criteria of the American Joint Committee on Cancer (Amin et al. 2017AMIN MB, GRESS DM, VEJA LRM, EDGE SB, GREENE FL, BYRD DR, BROOKLAND RK, WASHINGTON MK & COMPTON CC. 2017. AJCC Cancer Staging Manual. 8 th Ed. New York: Springer, 1032 p.). Tumor regression was evaluated according to the following criteria of the College of American Pathologists (Shon et al. 2021SHON W ET AL. 2021. Protocol for the Examination of Excision Specimens From Patients With Melanoma of the Skin. College of American Pathologists (CAP)., 2022): replacement of tumor cells by lymphocytic inflammation, as well as attenuation of epidermal and dermal fibrosis with inflammatory cells, melanophagocytosis and telangiectasia. The intensity of lymphocytic infiltration was assessed semi-quantitatively, and varied from 0 to 3, as follows: 0 = absent (no lymphocytic infiltration); 1 = mild (lymphocytes focally present in the peritumoral area without intratumoral extension); 2 = moderate (presence of lymphocytes in the peritumoral area with prominent intratumoral extension); 3 = marked (marked lymphocytic infiltration in the intra- and peritumoral areas) (Taube et al. 2014TAUBE JM, KLEIN A, BRAHMER JR, XU H, PAN X & KIM JH. 2014. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20: 5064-5074.).

Immunohistochemical (IHC) Analysis

The previously analyzed paraffin blocks of the H&E sections were selected in 3μm sections for the IHC analysis.

The monoclonal antibodies Peripheral Node Addressin (clone MECA-79 0.5mg, Novus Biological®) and anti-PDL-1 (clone E1L3N 0.1mg, Cell Signaling®) were used for the detection of high endothelial venules (HEV) and of PDL-1 expression. The slides were incubated overnight with the primary antibodies at room temperature in dilutions of 1:150 and 1:300, respectively. Controls positive for MECA-79 (palatine tonsils) and for PDL-1 (placenta) were included on all the slides. Immunostaining was processed using the chromogen diaminobenzidine (DAB) and all the slides were counterstained with Giemsa.

The IHC analysis was performed by two pathologists participating in this study, using a Zeiss Axio Scope.A1 optical microscope, and the photomicrographs were taken with a Zeiss AxioCam 5.0 camera.

Quantification of High Endothelial Venule Density

The absolute number of MECA-79-positive venules was quantified in the largest area of each tumor section. Venule density, expressed as the number of positive venules per mm2, was calculated semi-quantitatively for each case, based on previous studies (Martinet et al. 2012aMARTINET L, GARRIDO I & GIRARD JP. 2012b. Tumor high endothelial venules (HEVs) predict lymphocyte infiltration and favorable prognosis in breast cancer. Oncoimmunology 1: 789-790., Avram et al. 2013AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861.).

Evaluation of Pdl-1 Expression

PDL-1 expression was analyzed semi-quantitatively, based on the percentage (%) of labeled tumor cells (TC). Samples were considered positive when complete or incomplete cell membrane immunostaining of any intensity was observed in ≥ 1% of the TC. The following immunohistochemical scores (IHCS) were used for each immunohistochemical section: IHCS 0: < de 1% IHCS 1: ≥1% < 5% IHCS 2: ≥ 5% < 10%; IHCS 3: ≥ 10% (52). The intensity of membrane staining in TC was evaluated as weak (1), moderate (2) and intense (3). The location of PDL-1 positive TCs in relation to TILs was also analyzed.

Statistical analysis

The statistical analysis was performed using the SPSS 2.0 software package. The absolute and relative frequencies of the tumor’s clinicopathological characteristics were listed. The association of HEV density (MECA79 +) and PDL-1 expression with the clinicopathological variables was determined using the Chi-square test and the non-parametric Mann-Whitney and Kruskal-Wallis tests. The association between HEV density and PDL-1 expression was determined via the Kruskal-Wallis test. A p value of <0.05 was considered statistically significant.

RESULTS

The average age of the patients (60.3% women and 39.7% men) was 59.8 years. The most common site of primary melanoma was on the trunk (28.2%), followed by the lower limbs (24.4%), head and neck (20.5%) and upper limbs (20.5%). The predominant histological type was SSM (60.3%), followed by NM (17.9%), LMM (14.1%) and ALM (7.7%). Melanomas of the trunk were more frequent among males (63.6%), while melanomas of the lower limbs were more frequent among females (84.2%), with a statistical difference (p = 0.016). A comparison of the patient’s sex and type of melanoma revealed no statistical difference (p = 0.2). A higher rate of SSM (57.8%) was found among individuals younger than 55 years old. Most of the patients older than 55 had LMM (55.4%) and ALM (66.7%). Most of the tumors presented Clark levels II (30.8%) and III (38.5%), and Breslow thickness of up to 1mm (47.4%). Tumor infiltrating lymphocytes (TILs) were found in 94.9%, ulceration in 29.5% and regression signs in 19.2% of the patients.

MECA-79-positive venules were found in 69.2% (54/78) of the melanoma samples. The mean density of all MECA-79-positive venules per mm2 of tumor area was 2.38. Areas with higher HEV density also showed more prominent lymphocytic infiltrate (Fig. 1a,b).

Figure 1
IHC with MECA-79. a) Positive HEVs in the area of the tumor (100x magnification). b) Positive HEVs among TILs (400x magnification).

A positive association was found between HEV density and lymphocytic infiltration. This association was stronger with moderate (2) and severe (3) infiltrates, with significant differences in relation to absent or mild infiltrates (p <0.001) (Fig. 2). Higher HEV density was also associated with melanoma histological type, observed in SSM (2.59) and LMM (4.74), with a statistical difference (p <0.025). Melanomas with Breslow thickness of up to 1mm presented higher HEV density (3.7), with a statistical difference (p <0.004). Ulceration was observed in melanomas with a lower density of MECA-79-positive venules (p <0.003). No significant association was found between HEV density and Clark level, anatomical site of the lesion, presence of mitosis and regression signs. Table I summarizes the clinicopathological characteristics of melanomas and their association with HEV density.

Figure 2
Density of HEVs and TILs in primary cutaneous melanomas.
Table I
Association between clinicopathological characteristics and HEV density in primary cutaneous melanomas.

PDL-1 expression on TC was observed in 59% (46/78) of the specimens examined. When present, it was predominantly observed in association with TILs (Fig. 3a-d). This pattern was more conspicuous when the infiltrate intensity was moderate to severe than when it was mild or absent, and showed a significant association (p <0.007). Moreover, PDL-1 expression levels were higher the greater the intensity of lymphocytic infiltrate (Fig. 4). No significant association was found between PDL-1 expression levels and histological type, lesion site, Clark level, Breslow thickness, ulceration, regression signs and mitoses. It should be noted that, although there was no significant difference, thicker melanomas showed higher levels of PDL-1 expression and most of the acral melanomas were negative. Table II summarizes the association between PDL-1 expression and the pathological features of melanomas.

Figure 3
IHC with anti-PDL-1 antibody. a) Membrane positivity of PDL-1 in malignant melanocytes (3+ strong intensity), 40x magnification. b) Membrane positivity of PDL-1 in malignant melanocytes (2+ moderate intensity), 40x magnification. c) Membrane positivity of PDL-1 in malignant melanocytes (1+ weak intensity), 100x magnification. d) Colocation of PDL-1 positive melanocytes in geographic relation to TILs, 40x magnification.
Figure 4
PDL-1 and TILs in primary cutaneous melanomas.
Table II
Association between pathological characteristics and PDL-1 expression in primary cutaneous melanomas.

No association was found between PDL-1 expression and MECA-79-positive venule density (Fig. 5).

Figure 5
Association between HEV density and PDL-1 expression in primary cutaneous melanomas.

DISCUSSION

In Brazil, cutaneous melanoma occurs homogeneously in both sexes, but slightly more frequently among women, albeit without significant differences (Matheus & Verri 2015MATHEUS LGM & VERRI BHMA. 2015. Aspectos epidemiológicos do melanoma cutâneo epidemiological aspects of cutaneous melanoma. Rev Cienc Estud Acad Med 1(3).), a fact that was confirmed in this study. Moreover, individuals younger than 40 years of age rarely suffer from cutaneous melanoma. After the fourth decade of life, age-specific incidence rates increase, peaking in the seventh and eighth decades. As for primary tumor location, the preferential location among men is on the trunk and among women on the lower limbs (McCourt et al. 2014MCCOURT C, DOLAN O & GORMLEY G. 2014. Malignant melanoma: a pictorial review. Ulster Med J 83: 103-110., Matheus & Verri 2015MATHEUS LGM & VERRI BHMA. 2015. Aspectos epidemiológicos do melanoma cutâneo epidemiological aspects of cutaneous melanoma. Rev Cienc Estud Acad Med 1(3).). It is also known that age is associated with tumor location, with melanomas on the trunk occurring between the 5th and 6th decades, and head and neck melanomas occurring in patients in their 60s and 70s (Matheus & Verri 2015MATHEUS LGM & VERRI BHMA. 2015. Aspectos epidemiológicos do melanoma cutâneo epidemiological aspects of cutaneous melanoma. Rev Cienc Estud Acad Med 1(3).). In our study, the higher frequency of melanoma among women and of the SSM and NM types is consistent with previously published data, as is the finding that these tumors occur more commonly on the trunk, among men, and on the lower limbs, among women (McCourt et al. 2014MCCOURT C, DOLAN O & GORMLEY G. 2014. Malignant melanoma: a pictorial review. Ulster Med J 83: 103-110., Matheus & Verri 2015MATHEUS LGM & VERRI BHMA. 2015. Aspectos epidemiológicos do melanoma cutâneo epidemiological aspects of cutaneous melanoma. Rev Cienc Estud Acad Med 1(3)., Vazquez et al. 2015VAZQUEZ V, SILVA T, VIEIRA M, DE OLIVEIRA A, LISBOA M & DE ANDRADE D. 2015. Melanoma characteristics in Brazil: demographics, treatment, and survival analysis. BMC Res Notes 8: 4.).

The presence of TILs represents the host’s immune response against tumor cells. Numerous studies have drawn attention to a favorable prognosis of TILs in many solid tumors (Clemente et al. 1996CLEMENTE CG, MIHM MC, BUFALINO R, ZURRIDA S, COLLINI P & CASCINELLI N. 1996. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77: 1303-1310., Taylor et al. 2007TAYLOR RC, PATEL A, PANAGEAS KS, BUSAM KJ & BRADY MS. 2007. Tumor-Infiltrating Lymphocytes Predict Sentinel Lymph Node Positivity in Patients with Cutaneous Melanoma. J Clin Oncol 25: 869-875., Oble et al. 2009OBLE DA, LOEWE R, YU P & MIHM MC. 2009. Focus on TILs: Prognostic significance of tumor infiltrating lymphocytes in human melanoma. Cancer Immun 9: 1-20., Thomas et al. 2013THOMAS NE, BUSAM KJ, FROM L, KRICKER A, ARMSTRONG BK & ANTON-CULVER H. 2013. Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J Clin Oncol 31: 4252-4259., Fortes et al. 2015FORTES C, MASTROENI S, MANNOORANPARAMPIL TJ, PASSARELLI F, ZAPPALA A & ANNESSI G. 2015. Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res 25: 306-311., Santoiemma & Powell 2015SANTOIEMMA PP & POWELL DJ. 2015. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther 16: 807-820., Keun Park et al. 2017KEUN PARK C, KYUM KIM S, KEUN PARK C & KYUM KIM S. 2017. Clinicopathological significance of intratumoral and peritumoral lymphocytes and lymphocyte score based on the histologic subtypes of cutaneous melanoma. Oncotarget 8: 14759-14769., Shen et al. 2018SHEN M, WANG J & REN X. 2018. New insights into tumor-infiltrating B lymphocytes in breast cancer: Clinical impacts and regulatory mechanisms. Front Immunol 9: 470.). HEVs are specialized venules that facilitate lymphocyte recruitment and migration into melanomas and other tumors (Kiss et al. 2007KISS J, TIMAR J, SOMLAI B, GILDE K, FEJOS Z & GAUDI I. 2007. Association of microvessel density with infiltrating cells in human cutaneous malignant melanoma. Pathol Oncol Res 13: 21-31., Martinet et al. 2012aMARTINET L, GARRIDO I & GIRARD JP. 2012b. Tumor high endothelial venules (HEVs) predict lymphocyte infiltration and favorable prognosis in breast cancer. Oncoimmunology 1: 789-790., b, Avram et al. 2013AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861., Blanchard & Girard 2021BLANCHARD L & GIRARD JP. 2021. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 4: 719-753., Asrir et al. 2022ASRIR A ET AL. 2022. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell 3: 318-334.). Higher densities of these venules are linked with thinner melanomas, lower degrees of invasion and regression signs (Kiss et al. 2007KISS J, TIMAR J, SOMLAI B, GILDE K, FEJOS Z & GAUDI I. 2007. Association of microvessel density with infiltrating cells in human cutaneous malignant melanoma. Pathol Oncol Res 13: 21-31., Martinet et al. 2012aMARTINET L, GARRIDO I & GIRARD JP. 2012b. Tumor high endothelial venules (HEVs) predict lymphocyte infiltration and favorable prognosis in breast cancer. Oncoimmunology 1: 789-790., b, Avram et al. 2013AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861.).

Our findings confirm the hypothesis that HEVs are present in most cutaneous melanomas, and that their presence and density may predict a more effective immune response (Blanchard & Girard 2021BLANCHARD L & GIRARD JP. 2021. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 4: 719-753., Asrir et al. 2022ASRIR A ET AL. 2022. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell 3: 318-334.), given the positive association between histopathological variables with a better prognosis and the presence and degree of TILs in tumors with a higher density of these venules. Moreover, it seems reasonable to propose the idea that HEVs can recruit TILs and facilitate their migration into these tumors. A previous study found no significant association between HEV density and the anatomical location of the tumor, a finding that was confirmed in our study (Avram et al. 2013AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861.). The higher HEV density found in some histological subtypes in this study (higher in LMM and lower in ALM) is in agreement with previously described findings (Weinstein & Storkus 2016WEINSTEIN AM & STORKUS WJ. 2016. Biosynthesis and functional significance of peripheral node addressin in cancer-associated TLO. Front Immunol 7: 1-8.). Therefore, the higher HEV density found in LMMs and in thinner melanomas (<1mm) with a higher degree of TILs corroborates our proposition that these vessels constitute a good prognostic biomarker.

There are also mechanisms of tumor immune evasion, an important feature of some tumors, which affect tumor progression and survival. Thus, PDL-1 expression on tumor cells and consequent binding to the PD-1 receptor in lymphocytes leads to their inactivation, reducing the chance of antitumor immune response. Melanomas that express PDL-1 present shorter survival and cells that express this marker are geographically located adjacent to tumor infiltrating lymphocytes (Kashani-Sabet 2010KASHANI-SABET M. 2010. Tumor progression by immune evasion in melanoma: Role of the programmed cell death-1/programmed cell death-1 ligand 1 interaction. Cancer 116: 1623-1625., Taube et al. 2013TAUBE JM, ANDERS RA, YOUNG GD, XU H, SHARMA R & MCMILLER TL. 2013. Mechanism of Immune Escape. Sci Transl Med 4: 1-22., Massi et al. 2014MASSI D, BRUSA D, MERELLI B, CIANO M, AUDRITO V & SERRA S. 2014. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann Oncol 25: 2433-2442., Jin et al. 2015JIN Y, ZAHO J, SHI X & YU X. 2015. Prognostic value of programed death ligand 1 in patients with solid tumors: A meta-analysis. J Cancer Res Ther 11: C38-C43., Madore et al. 2015MADORE J, VILAIN RE, MENZIES AM, KAKAVAND H, WILMOTT JS & HYMAN J. 2015. PD-L1 expression in melanoma shows marked heterogeneity within and between patients: Implications for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res 28: 245-253.). Presumably, therefore, treatments aimed at blocking this PD-1/PDL-1 axis may offer clinical benefits by reactivating the antitumor immune response and preventing tumor progression, reducing the likelihood of metastasis. Studies involving the use of antibodies that block the interaction of PD-1 with its ligands in patients with advanced melanomas have proved to be clinically beneficial (Sapoznik et al. 2012SAPOZNIK S, HAMMER O, ORTENBERG R, BESSER MJ, BEN-MOSHE T & SCHACHTER J. 2012. Novel anti-melanoma immunotherapies: Disarming tumor escape mechanisms. Clin Dev Immunol 2012: 818214., Tsai et al. 2014TSAI KK, ZARZOSO I & DAUD AI. 2014. PD-1 and PD-L1 antibodies for melanoma. Hum Vaccin Immunother 10: 3111-3116., Ascierto & Marincola 2015ASCIERTO PA & MARINCOLA FM. 2015. EBioMedicine 2015: The Year of Anti-PD-1 / PD-L1s Against Melanoma and Beyond. EBIOM 2: 92-93., Carbognin et al. 2015CARBOGNIN L, PILOTTO S, MILELLA M, VACCARO V & BRUNELLI M. 2015. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourin Cancers. PLoS ONE 10(6): e0130142., Philips & Atkins 2015PHILIPS GK & ATKINS M. 2015. Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Int Immunol 27: 39-46.).

Our findings demonstrate that the expression of PDL-1, in most cases found to occur adjacent to TILs, may be a mechanism of tumor immune evasion, since the hyperexpression of this ligand and consequent binding to its PD-1 receptor on lymphocytes leads to their inactivation. Melanomas with greater PDL-1 expression are more heterogeneous and belong to a group of diseases with shorter survival rates, with greater chances of recurrence and metastases (Gadiot et al. 2011GADIOT J, HOOIJKAAS AI, KAISER ADM, VAN TINTEREN H, VAN BOVEN H & BLANK C. 2011. Overall survival and PD-L1 expression in metastasized malignant melanoma. Cancer 117: 2192-2201., Massi et al. 2014MASSI D, BRUSA D, MERELLI B, CIANO M, AUDRITO V & SERRA S. 2014. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann Oncol 25: 2433-2442., Muenst et al. 2014MUENST S, SCHAERLI AR, GAO F, DUSTER S, TRELLA E & DROESER RA. 2014. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 146: 15-24., Xu et al. 2015XU F, XU L, WANG Q, AN G, FENG G & LIU F. 2015. Clinicopathological and prognostic value of programmed death ligand-1 (PD-L1) in renal cell carcinoma: a meta-analysis. Int J Clin Exp Med 8: 14595-14603.). Our findings also indicate that tumors that express PDL-1 are thicker melanomas with a higher degree of TILs, confirming that PDL-1 expression in geographical relation to TILs may be important mechanism of attempted immune evasion. Although studies have described the negative impact on tumor progression and survival of melanomas with higher PDL-1 expression, patients with this profile are candidates for immunotherapy aimed at blocking this axis, with good response rates to these treatments and with clinical benefits (McCourt et al. 2014MCCOURT C, DOLAN O & GORMLEY G. 2014. Malignant melanoma: a pictorial review. Ulster Med J 83: 103-110., Velcheti et al. 2014VELCHETI V, SCHALPER KA, CARVAJAL DE, ANAGNOSTOU VK, SYRIGOS KN & SZNOL M. 2014. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Investig 94: 107-116., Herbst et al. 2014HERBST RS, SORIA J, KOWANETZ M, FINE GD, HAMID O & KOHRT HEK. 2014. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563-567., Matheus & Verri 2015MATHEUS LGM & VERRI BHMA. 2015. Aspectos epidemiológicos do melanoma cutâneo epidemiological aspects of cutaneous melanoma. Rev Cienc Estud Acad Med 1(3).). In addition, the combination of angiogenic and anti-PDL-1 therapies can stimulate antitumor immunity through the formation of HEVs, making the study of these two biomarkers of tumor immune response and their correlation particularly interesting (Hayasaka et al. 2010HAYASAKA H, TANIGUCHI K, FUKAI S & MIYASAKA M. 2010. Neogenesis and development of the high endothelial venules that mediate lymphocyte trafficking. Cancer Sci 101: 2302-2308., Allen et al. 2017ALLEN E, JABOUILLE A, RIVERA LB, LODEWIJCKX I, MISSIAEN R & STERI V. 2017. Combined antiangiogenic and anti–PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med 9.).

In conclusion, the identification and quantification of HEVs, the determination of PDL-1 expression and its association with the presence and degree of TILs provides more information about the histopathological characteristics and prognosis of primary cutaneous melanomas. The findings garnered in this study are of great interest, since they may underpin further studies aimed at developing new therapeutic strategies combining the stimulation of HEV formation with blockage of the PD-1/PDL-1 axis. This may boost the antitumor immune response by facilitating the migration of TILs and the deactivation of the tumor immune evasion mechanism.

ACKNOWLEDGMENTS

We are indebted to the Institute of Pathology of Passo Fundo, and particularly to its employees Cristiane Trentim, Jaime Baril, Sidnei Abido, Marcia Pitol and Tabata Bedin, for their help in selecting samples from the biobank and in performing the immunohistochemistry tests.

REFERENCES

  • ALLEN E, JABOUILLE A, RIVERA LB, LODEWIJCKX I, MISSIAEN R & STERI V. 2017. Combined antiangiogenic and anti–PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med 9.
  • AMIN MB, GRESS DM, VEJA LRM, EDGE SB, GREENE FL, BYRD DR, BROOKLAND RK, WASHINGTON MK & COMPTON CC. 2017. AJCC Cancer Staging Manual. 8 th Ed. New York: Springer, 1032 p.
  • ASCIERTO PA & MARINCOLA FM. 2015. EBioMedicine 2015: The Year of Anti-PD-1 / PD-L1s Against Melanoma and Beyond. EBIOM 2: 92-93.
  • ASRIR A ET AL. 2022. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell 3: 318-334.
  • AVRAM G, SÁNCHEZ-SENDRA B, MARTÍN JM, TERRÁDEZ L, RAMOS D & MONTEAGUDO C. 2013. The density and type of MECA-79-positive high endothelial venules correlate with lymphocytic infiltration and tumour regression in primary cutaneous melanoma. Histopathology 63: 852-861.
  • AZIMI F, SCOLYER RA, RUMCHEVA P, MONCRIEFF M, MURALI R & MCCARTHY SW. 2012. Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol 30: 2678-2683.
  • BECKERS RK, SELINGER CI, VILAIN R, MADORE J, WILMOTT JS & HARVEY K. 2016. Programmed death ligand 1 expression in triple-negative breast cancer is associated with tumour-infiltrating lymphocytes and improved outcome. Histopathology 69: 25-34.
  • BLANCHARD L & GIRARD JP. 2021. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 4: 719-753.
  • CARBOGNIN L, PILOTTO S, MILELLA M, VACCARO V & BRUNELLI M. 2015. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourin Cancers. PLoS ONE 10(6): e0130142.
  • CHEN R, PENG PC, WEN B, LI FY, XIE S & CHEN G. 2016. Anti–Programmed Cell Death (PD)-1 Immunotherapy for Malignant Tumor: A Systematic Review and Meta-Analysis. Transl Oncol 9: 32-40.
  • CLEMENTE CG, MIHM MC, BUFALINO R, ZURRIDA S, COLLINI P & CASCINELLI N. 1996. Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77: 1303-1310.
  • DAUD AI, WOLCHOK JD, ROBERT C, HWU WJ, WEBER JS & RIBAS A. 2016. Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol 34: 4102-4109.
  • DRAYTON DL, YING X, LEE J, LESSLAUER W & RUDDLE NH. 2003. Ectopic LTO: Directs Lymphoid Organ Neogenesis with Concomitant Expression of Peripheral Node Addressin and a HEV-restricted Sulfotransferase. J Experiment Med 197(9): 1153-1163.
  • DUPRAT JP, BRECHTBÜLH ER, COSTA DE SÁ B, ENOKIHARA M, FREGNANI JH & LANDMAN G. 2016. Absence of Tumor-Infiltrating Lymphocyte Is a Reproducible Predictive Factor for Sentinel Lymph Node Metastasis: A Multicenter Database Study by the Brazilian Melanoma Group. PLoS ONE 11 : e0148160.
  • FORTES C, MASTROENI S, MANNOORANPARAMPIL TJ, PASSARELLI F, ZAPPALA A & ANNESSI G. 2015. Tumor-infiltrating lymphocytes predict cutaneous melanoma survival. Melanoma Res 25: 306-311.
  • GADIOT J, HOOIJKAAS AI, KAISER ADM, VAN TINTEREN H, VAN BOVEN H & BLANK C. 2011. Overall survival and PD-L1 expression in metastasized malignant melanoma. Cancer 117: 2192-2201.
  • GALLIMORE A, GODKIN A & AGER A. 2013. High Endothelial Venules, Help or hindrance in the quest for antitumor immunity? Oncoimmunology 2(5): e24272.
  • HASAN A, GHEBEH H, LEHE C, AHMAD R & DERMIME S. 2011. Therapeutic targeting of B7-H1 in breast cancer. Expert Opin Ther Targets 15: 1211-1225.
  • HAYASAKA H, TANIGUCHI K, FUKAI S & MIYASAKA M. 2010. Neogenesis and development of the high endothelial venules that mediate lymphocyte trafficking. Cancer Sci 101: 2302-2308.
  • HEMMERICH S, BUTCHER EC & ROSEN SD. 1994. Sulfation-dependent recognition of high endothelial venules (HEV)-ligands by L-selectin and MECA 79, and adhesion-blocking monoclonal antibody. J Exp Med 180: 2219-2226.
  • HERBST RS, SORIA J, KOWANETZ M, FINE GD, HAMID O & KOHRT HEK. 2014. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563-567.
  • HINO R, KABASHIMA K, KATO Y, YAGI H, NAKAMURA M & HONJO T. 2010. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 116: 1757-1766.
  • JIN Y, ZAHO J, SHI X & YU X. 2015. Prognostic value of programed death ligand 1 in patients with solid tumors: A meta-analysis. J Cancer Res Ther 11: C38-C43.
  • JUNEJA VR, MCGUIRE KA, MANGUSO RT, LAFLEUR MW, COLLINS N & HAINING WN. 2017. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med 214: 895-904.
  • KASHANI-SABET M. 2010. Tumor progression by immune evasion in melanoma: Role of the programmed cell death-1/programmed cell death-1 ligand 1 interaction. Cancer 116: 1623-1625.
  • KEUN PARK C, KYUM KIM S, KEUN PARK C & KYUM KIM S. 2017. Clinicopathological significance of intratumoral and peritumoral lymphocytes and lymphocyte score based on the histologic subtypes of cutaneous melanoma. Oncotarget 8: 14759-14769.
  • KISS J, TIMAR J, SOMLAI B, GILDE K, FEJOS Z & GAUDI I. 2007. Association of microvessel density with infiltrating cells in human cutaneous malignant melanoma. Pathol Oncol Res 13: 21-31.
  • LEITE KR, REIS ST, JUNIOR JP, ZERATI M, GOMES DDE O & CAMARA-LOPES LH. 2015. PD-L1 expression in renal cell carcinoma clear cell type is related to unfavorable prognosis. Diagn Pathol 10: 189.
  • MADORE J, VILAIN RE, MENZIES AM, KAKAVAND H, WILMOTT JS & HYMAN J. 2015. PD-L1 expression in melanoma shows marked heterogeneity within and between patients: Implications for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res 28: 245-253.
  • MAIBACH F, SADOZAI H, SEYED JAFARI SM, HUNGER RE & SCHENK M. 2020. Tumor-infiltrating lymphocytes and their prognostic value in cutaneous melanoma. Frontiers Immunol 11: 2105.
  • MARTINET L, GARRIDO I, FILLERON T, GUELLEC S LE, BELLARD E & FOURNIE JJ. 2011. Human solid tumors contain high endothelial venules: Association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res 71: 5678-5687.
  • MARTINET L, GARRIDO I & GIRARD JP. 2012b. Tumor high endothelial venules (HEVs) predict lymphocyte infiltration and favorable prognosis in breast cancer. Oncoimmunology 1: 789-790.
  • MARTINET L, LE GUELLEC S, FILLERON T, LAMANT L, MEYER N & ROCHAIX P. 2012a. High endothelial venules (HEVs) in human melanoma lesions: Major gateways for tumor-infiltrating lymphocytes. Oncoimmunology 1: 829-839.
  • MASSI D, BRUSA D, MERELLI B, CIANO M, AUDRITO V & SERRA S. 2014. PD-L1 marks a subset of melanomas with a shorter overall survival and distinct genetic and morphological characteristics. Ann Oncol 25: 2433-2442.
  • MATHEUS LGM & VERRI BHMA. 2015. Aspectos epidemiológicos do melanoma cutâneo epidemiological aspects of cutaneous melanoma. Rev Cienc Estud Acad Med 1(3).
  • MCCOURT C, DOLAN O & GORMLEY G. 2014. Malignant melanoma: a pictorial review. Ulster Med J 83: 103-110.
  • MICHIE SA, STREETER PR, BOLT PA, BUTCHER EC & PICKER LJ. 1993. The Human Peripheral Lymph Node Vascular Addressin An Inducible Endothelial Antigen Involved in Lymphocyte Homing. Am J Pathol 143: 1688-1698.
  • MIDDLETON J, AMERICH L, GAYON R, JULIEN D, MANSAT M & MANSAT P. 2005. A comparative study of endothelial cell markers expressed in chronically inflamed human tissues: MECA-79, Duffy antigen receptor for chemokines, von Willebrand factor, CD31, CD34, CD105 and CD146. J Pathol 206: 260-268.
  • MIYASAKA M & TANAKA T. 2004. Lymphocyte trafficking across high endothelial venules: dogmas and enigmas. Nat Rev Immunol 1: 360.
  • MUENST S, SCHAERLI AR, GAO F, DUSTER S, TRELLA E & DROESER RA. 2014. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 146: 15-24.
  • OBLE DA, LOEWE R, YU P & MIHM MC. 2009. Focus on TILs: Prognostic significance of tumor infiltrating lymphocytes in human melanoma. Cancer Immun 9: 1-20.
  • PERIVOLIOTIS K, NTELLAS P, DADOULI K, KOUTOUKOGLOU P, IOANNOU M & TEPETES K. 2017. Microvessel Density in Patients with Cutaneous Melanoma: An Up-to-Date Systematic Review and Meta-Analysis. J Skin Cancer 2017.
  • PHILIPS GK & ATKINS M. 2015. Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Int Immunol 27: 39-46.
  • QUAIL DF & JOYCE JA. 2013. Microenvironmental regulation of tumor progression and metastasis. Nat Med 19: 1423-1437.
  • SANLORENZO M, VUJIC I, POSCH C, DAJEE A, YEN A & KIM S. 2014. Melanoma immunotherapy. Cancer Biol Ther 15: 665-674.
  • SANTOIEMMA PP & POWELL DJ. 2015. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther 16: 807-820.
  • SAPOZNIK S, HAMMER O, ORTENBERG R, BESSER MJ, BEN-MOSHE T & SCHACHTER J. 2012. Novel anti-melanoma immunotherapies: Disarming tumor escape mechanisms. Clin Dev Immunol 2012: 818214.
  • SCHALPER KA. 2014. PD-L1 expression and tumor-infiltrating lymphocytes: Revisiting the antitumor immune response potential in breast cancer. Oncoimmunology 3: e29288.
  • SHEN M, WANG J & REN X. 2018. New insights into tumor-infiltrating B lymphocytes in breast cancer: Clinical impacts and regulatory mechanisms. Front Immunol 9: 470.
  • SHON W ET AL. 2021. Protocol for the Examination of Excision Specimens From Patients With Melanoma of the Skin. College of American Pathologists (CAP).
  • SHON W, NAGARAJAN P, FRISHBERG DP, GERSHENWALD J, FLOTTE MD, MCCALMONT TH & SMOLLER BR. 2023. Protocol for the Examination of Biopsy Specimens From Patients With Invasive Melanoma of the Skin. College of American Pathologists (CAP).
  • SINHA RK, YANG G, ALEXANDER C & MAGE RG. 2006. De novo expression of MECA-79 glycoprotein-determinant on developing B lymphocytes in gut-associated lymphoid tissues. Immunology 119: 461-469.
  • TAUBE JM, ANDERS RA, YOUNG GD, XU H, SHARMA R & MCMILLER TL. 2013. Mechanism of Immune Escape. Sci Transl Med 4: 1-22.
  • TAUBE JM, KLEIN A, BRAHMER JR, XU H, PAN X & KIM JH. 2014. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20: 5064-5074.
  • TAYLOR RC, PATEL A, PANAGEAS KS, BUSAM KJ & BRADY MS. 2007. Tumor-Infiltrating Lymphocytes Predict Sentinel Lymph Node Positivity in Patients with Cutaneous Melanoma. J Clin Oncol 25: 869-875.
  • THOMAS NE, BUSAM KJ, FROM L, KRICKER A, ARMSTRONG BK & ANTON-CULVER H. 2013. Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J Clin Oncol 31: 4252-4259.
  • TSAI KK, ZARZOSO I & DAUD AI. 2014. PD-1 and PD-L1 antibodies for melanoma. Hum Vaccin Immunother 10: 3111-3116.
  • UCHIMURA K & ROSEN SD. 2006. Sulfated L-selectin ligands as a therapeutic target in chronic inflammation. Trends Immunol 27: 559-565.
  • VAZQUEZ V, SILVA T, VIEIRA M, DE OLIVEIRA A, LISBOA M & DE ANDRADE D. 2015. Melanoma characteristics in Brazil: demographics, treatment, and survival analysis. BMC Res Notes 8: 4.
  • VELCHETI V, SCHALPER KA, CARVAJAL DE, ANAGNOSTOU VK, SYRIGOS KN & SZNOL M. 2014. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Investig 94: 107-116.
  • WEINSTEIN AM & STORKUS WJ. 2016. Biosynthesis and functional significance of peripheral node addressin in cancer-associated TLO. Front Immunol 7: 1-8.
  • WIMBERLY H, BROWN JR, SCHALPER K, HAACK H, SILVER MR & NIXON C. 2015. PD-L1 Expression Correlates with Tumor-Infiltrating Lymphocytes and Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancer Immunol Res 3: 326-332.
  • WU P, WU D, LI L, CHAI Y & HUANG J. 2015. PD-L1 and survival in solid tumors: A meta-analysis. PLoS ONE 10: 1-15.
  • XU F, XU L, WANG Q, AN G, FENG G & LIU F. 2015. Clinicopathological and prognostic value of programmed death ligand-1 (PD-L1) in renal cell carcinoma: a meta-analysis. Int J Clin Exp Med 8: 14595-14603.
  • ZENG J, ZHANG XK, CHEN HD, ZHONG ZH, WU QL & LIN SX. 2016. Expression of programmed cell death-ligand 1 and its correlation with clinical outcomes in gliomas. Oncotarget 7(8): 8944.

Publication Dates

  • Publication in this collection
    18 Mar 2024
  • Date of issue
    2024

History

  • Received
    14 Apr 2023
  • Accepted
    17 July 2023
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br