#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Quantitative Measurement of Melanoma Spread in Sentinel Lymph Nodes and Survival


Background:
Sentinel lymph node spread is a crucial factor in melanoma outcome. We aimed to define the impact of minimal cancer spread and of increasing numbers of disseminated cancer cells on melanoma-specific survival.

Methods and Findings:
We analyzed 1,834 sentinel nodes from 1,027 patients with ultrasound node-negative melanoma who underwent sentinel node biopsy between February 8, 2000, and June 19, 2008, by histopathology including immunohistochemistry and quantitative immunocytology. For immunocytology we recorded the number of disseminated cancer cells (DCCs) per million lymph node cells (DCC density [DCCD]) after disaggregation and immunostaining for the melanocytic marker gp100. None of the control lymph nodes from non-melanoma patients (n = 52) harbored gp100-positive cells. We analyzed gp100-positive cells from melanoma patients by comparative genomic hybridization and found, in 45 of 46 patients tested, gp100-positive cells displaying genomic alterations. At a median follow-up of 49 mo (range 3–123 mo), 138 patients (13.4%) had died from melanoma. Increased DCCD was associated with increased risk for death due to melanoma (univariable analysis; p<0.001; hazard ratio 1.81, 95% CI 1.61–2.01, for a 10-fold increase in DCCD + 1). Even patients with a positive DCCD ≤3 had an increased risk of dying from melanoma compared to patients with DCCD = 0 (p = 0.04; hazard ratio 1.63, 95% CI 1.02–2.58). Upon multivariable testing DCCD was a stronger predictor of death than histopathology. The final model included thickness, DCCD, and ulceration (all p<0.001) as the most relevant prognostic factors, was internally validated by bootstrapping, and provided superior survival prediction compared to the current American Joint Committee on Cancer staging categories.

Conclusions:
Cancer cell dissemination to the sentinel node is a quantitative risk factor for melanoma death. A model based on the combined quantitative effects of DCCD, tumor thickness, and ulceration predicted outcome best, particularly at longer follow-up. If these results are validated in an independent study, establishing quantitative immunocytology in histopathological laboratories may be useful clinically.

Please see later in the article for the Editors' Summary


Vyšlo v časopise: Quantitative Measurement of Melanoma Spread in Sentinel Lymph Nodes and Survival. PLoS Med 11(2): e32767. doi:10.1371/journal.pmed.1001604
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pmed.1001604

Souhrn

Background:
Sentinel lymph node spread is a crucial factor in melanoma outcome. We aimed to define the impact of minimal cancer spread and of increasing numbers of disseminated cancer cells on melanoma-specific survival.

Methods and Findings:
We analyzed 1,834 sentinel nodes from 1,027 patients with ultrasound node-negative melanoma who underwent sentinel node biopsy between February 8, 2000, and June 19, 2008, by histopathology including immunohistochemistry and quantitative immunocytology. For immunocytology we recorded the number of disseminated cancer cells (DCCs) per million lymph node cells (DCC density [DCCD]) after disaggregation and immunostaining for the melanocytic marker gp100. None of the control lymph nodes from non-melanoma patients (n = 52) harbored gp100-positive cells. We analyzed gp100-positive cells from melanoma patients by comparative genomic hybridization and found, in 45 of 46 patients tested, gp100-positive cells displaying genomic alterations. At a median follow-up of 49 mo (range 3–123 mo), 138 patients (13.4%) had died from melanoma. Increased DCCD was associated with increased risk for death due to melanoma (univariable analysis; p<0.001; hazard ratio 1.81, 95% CI 1.61–2.01, for a 10-fold increase in DCCD + 1). Even patients with a positive DCCD ≤3 had an increased risk of dying from melanoma compared to patients with DCCD = 0 (p = 0.04; hazard ratio 1.63, 95% CI 1.02–2.58). Upon multivariable testing DCCD was a stronger predictor of death than histopathology. The final model included thickness, DCCD, and ulceration (all p<0.001) as the most relevant prognostic factors, was internally validated by bootstrapping, and provided superior survival prediction compared to the current American Joint Committee on Cancer staging categories.

Conclusions:
Cancer cell dissemination to the sentinel node is a quantitative risk factor for melanoma death. A model based on the combined quantitative effects of DCCD, tumor thickness, and ulceration predicted outcome best, particularly at longer follow-up. If these results are validated in an independent study, establishing quantitative immunocytology in histopathological laboratories may be useful clinically.

Please see later in the article for the Editors' Summary


Zdroje

1. MortonDL, WenDR, WongJH, EconomouJS, CagleLA, et al. (1992) Technical details of intraoperative lymphatic mapping for early stage melanoma. Arch Surg 127: 392–399.

2. ReintgenD, CruseCW, WellsK, BermanC, FenskeN, et al. (1994) The orderly progression of melanoma nodal metastases. Ann Surg 220: 759–767.

3. GershenwaldJE, SoongSJ, BalchCM (2010) 2010 TNM staging system for cutaneous melanoma…and beyond. Ann Surg Oncol 17: 1475–1477.

4. BalchCM, GershenwaldJE, SoongSJ, ThompsonJF, AtkinsMB, et al. (2009) Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 27: 6199–6206.

5. MuraliR, DesilvaC, McCarthySW, ThompsonJF, ScolyerRA (2012) Sentinel lymph nodes containing very small (<0.1 mm) deposits of metastatic melanoma cannot be safely regarded as tumor-negative. Ann Surg Oncol 19: 1089–1099.

6. van AkkooiAC, de WiltJH, VerhoefC, SchmitzPI, van GeelAN, et al. (2006) Clinical relevance of melanoma micrometastases (<0.1 mm) in sentinel nodes: are these nodes to be considered negative? Ann Oncol 17: 1578–1585.

7. van der PloegAP, van AkkooiAC, SchmitzPI, KoljenovicS, VerhoefC, et al. (2010) EORTC Melanoma Group sentinel node protocol identifies high rate of submicrometastases according to Rotterdam criteria. Eur J Cancer 46: 2414–2421.

8. van DiestPJ (1999) Histopathological workup of sentinel lymph nodes: how much is enough? J Clin Pathol 52: 871–873.

9. AbrahamsenHN, Hamilton-DutoitSJ, LarsenJ, SteinicheT (2004) Sentinel lymph nodes in malignant melanoma: extended histopathologic evaluation improves diagnostic precision. Cancer 100: 1683–1691.

10. CochranAJ, BaldaBR, StarzH, BachterD, KragDN, et al. (2000) The Augsburg Consensus. Techniques of lymphatic mapping, sentinel lymphadenectomy, and completion lymphadenectomy in cutaneous malignancies. Cancer 89: 236–241.

11. StarzH, BaldaBR, KramerKU, BuchelsH, WangH (2001) A micromorphometry-based concept for routine classification of sentinel lymph node metastases and its clinical relevance for patients with melanoma. Cancer 91: 2110–2121.

12. MuraliR, ThompsonJF, ScolyerRA (2010) Location of melanoma metastases in sentinel lymph nodes: what are the implications for histologic processing of sentinel lymph nodes in routine practice? Am J Surg Pathol 34: 127–129.

13. BagariaSP, FariesMB, MortonDL (2010) Sentinel node biopsy in melanoma: technical considerations of the procedure as performed at the John Wayne Cancer Institute. J Surg Oncol 101: 669–676.

14. MortonDL, ThompsonJF, CochranAJ, MozzilloN, ElashoffR, et al. (2006) Sentinel-node biopsy or nodal observation in melanoma. N Engl J Med 355: 1307–1317.

15. PrietoVG, ClarkSH (2002) Processing of sentinel lymph nodes for detection of metastatic melanoma. Ann Diagn Pathol 6: 257–264.

16. ScolyerRA, MuraliR, McCarthySW, ThompsonJF (2008) Pathologic examination of sentinel lymph nodes from melanoma patients. Semin Diagn Pathol 25: 100–111.

17. UlmerA, FischerJR, SchanzS, SotlarK, BreuningerH, et al. (2005) Detection of melanoma cells displaying multiple genomic changes in histopathologically negative sentinel lymph nodes. Clin Cancer Res 11: 5425–5432.

18. KleinCA, BlankensteinTJ, Schmidt-KittlerO, PetronioM, PolzerB, et al. (2002) Genetic heterogeneity of single disseminated tumour cells in minimal residual cancer. Lancet 360: 683–689.

19. KleinCA, Schmidt-KittlerO, SchardtJA, PantelK, SpeicherMR, et al. (1999) Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells. Proc Natl Acad Sci U S A 96: 4494–4499.

20. BaudisM, ClearyML (2001) Progenetix.net: an online repository for molecular cytogenetic aberration data. Bioinformatics 17: 1228–1229.

21. Kalbfleisch JD, Prentice RL (1980) The statistical analysis of failure time data. Hoboken (New Jersey): Wiley. 321 p.

22. SchwarzG (1978) Estimating the dimension of a model. Ann Statist 6: 461–464.

23. HarrellFEJr, LeeKL, MarkDB (1996) Multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat Med 15: 361–387.

24. Otto E (1963) Nomography. New York: Macmillan.

25. McShaneLM, AltmanDG, SauerbreiW, TaubeSE, GionM, et al. (2005) Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst 97: 1180–1184.

26. MachadoSG, RobinsonGA (1994) A direct, general approach based on isobolograms for assessing the joint action of drugs in pre-clinical experiments. Stat Med 13: 2289–2309.

27. ScheriRP, EssnerR, TurnerRR, YeX, MortonDL (2007) Isolated tumor cells in the sentinel node affect long-term prognosis of patients with melanoma. Ann Surg Oncol 14: 2861–2866.

28. BraumullerH, WiederT, BrennerE, AssmannS, HahnM, et al. (2013) T-helper-1-cell cytokines drive cancer into senescence. Nature 494: 361–365.

29. KleinCA (2011) Framework models of tumor dormancy from patient-derived observations. Curr Opin Genet Dev 21: 42–49.

30. PeinadoH, AleckovicM, LavotshkinS, MateiI, Costa-SilvaB, et al. (2012) Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 18: 883–891.

31. PrietoVG (2012) Cutaneous melanocytic lesions: do not miss the invisible gorilla. Adv Anat Pathol 19: 263–269.

32. Riber-HansenR, NyengaardJR, Hamilton-DutoitSJ, SteinicheT (2009) The nodal location of metastases in melanoma sentinel lymph nodes. Am J Surg Pathol 33: 1522–1528.

33. MocellinS, HoonDS, PilatiP, RossiCR, NittiD (2007) Sentinel lymph node molecular ultrastaging in patients with melanoma: a systematic review and meta-analysis of prognosis. J Clin Oncol 25: 1588–1595.

34. SchillingD, HennenlotterJ, SotlarK, KuehsU, SengerE, et al. (2011) Quantification of tumor cell burden by analysis of single cell lymph node disaggregates in metastatic prostate cancer. Prostate 70: 1110–1118.

35. CzyzTZ, HoffmannM, SchlimokG, PolzerB, KleinCA (2004) Reliable single cell array CGH for clinical samples. PLoS ONE 9: e85907.

Štítky
Interné lekárstvo

Článok vyšiel v časopise

PLOS Medicine


2014 Číslo 2
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#