Somatic LKB1 mutations promote cervical cancer progression

Shana N Wingo, Teresa D Gallardo, Esra A Akbay, Mei-Chi Liang, Cristina M Contreras, Todd Boren, Takeshi Shimamura, David S Miller, Norman E Sharpless, Nabeel Bardeesy, David J Kwiatkowski, John O Schorge, Kwok-Kin Wong, Diego H Castrillon, Shana N Wingo, Teresa D Gallardo, Esra A Akbay, Mei-Chi Liang, Cristina M Contreras, Todd Boren, Takeshi Shimamura, David S Miller, Norman E Sharpless, Nabeel Bardeesy, David J Kwiatkowski, John O Schorge, Kwok-Kin Wong, Diego H Castrillon

Abstract

Human Papilloma Virus (HPV) is the etiologic agent for cervical cancer. Yet, infection with HPV is not sufficient to cause cervical cancer, because most infected women develop transient epithelial dysplasias that spontaneously regress. Progression to invasive cancer has been attributed to diverse host factors such as immune or hormonal status, as no recurrent genetic alterations have been identified in cervical cancers. Thus, the pressing question as to the biological basis of cervical cancer progression has remained unresolved, hampering the development of novel therapies and prognostic tests. Here we show that at least 20% of cervical cancers harbor somatically-acquired mutations in the LKB1 tumor suppressor. Approximately one-half of tumors with mutations harbored single nucleotide substitutions or microdeletions identifiable by exon sequencing, while the other half harbored larger monoallelic or biallelic deletions detectable by multiplex ligation probe amplification (MLPA). Biallelic mutations were identified in most cervical cancer cell lines; HeLa, the first human cell line, harbors a homozygous 25 kb deletion that occurred in vivo. LKB1 inactivation in primary tumors was associated with accelerated disease progression. Median survival was only 13 months for patients with LKB1-deficient tumors, but >100 months for patients with LKB1-wild type tumors (P = 0.015, log rank test; hazard ratio = 0.25, 95% CI = 0.083 to 0.77). LKB1 is thus a major cervical tumor suppressor, demonstrating that acquired genetic alterations drive progression of HPV-induced dysplasias to invasive, lethal cancers. Furthermore, LKB1 status can be exploited clinically to predict disease recurrence.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Somatic mutations and deletions of…
Figure 1. Somatic mutations and deletions of LKB1 in cervical tumors.
(A) Representative chromatograms of primary tumors. (B) C4I cell line. Lower panels, control DNA samples from each patient (for C4I, human peripheral leukocyte DNA). Wild-type sequences are shown below. Chromatograms represent forward strand except case #41 where reverse complement is shown to more clearly illustrate the deletion. Mutations are heterozygous except where indicated. (C) LKB1 deletions in primary cervical tumors by MLPA. Bars = relative signal intensity per probe. Sixteen probes (black) correspond to LKB1 locus on chromosome 19. Probe identifiers shown below. Probes 0.9M5′ and 0.6M5′ are ∼900 and 600 kb 5′ of locus (telomeric), while 10M3′ is ∼10000 kb 3′ (centromeric); remaining 13 probes correspond to LKB1 noncoding/coding exons. White bars correspond to randomly selected probes from other chromosomes.
Figure 2. LKB1 mutations occur in each…
Figure 2. LKB1 mutations occur in each of the principal histologic subtypes of cervical cancer.
(A) Histology of representative cases with LKB1 mutations (SCC = squamous cell carcinoma; Adeno = adenocarcinoma; AdenoSq = adenosquamous carcinoma; MDA = minimal deviation adenocarcinoma). Scale bar = 20 microns. (B) Relative distribution of the three principal histologic subtypes among LKB1-mutant (red) vs. LKB1-wild type (black) cases (totals = 100%) shows that the histologic spectrum is virtually identical in LKB1 null vs. wild-type tumors.
Figure 3. Homozygous LKB1 deletions occur in…
Figure 3. Homozygous LKB1 deletions occur in majority of cervical cancer cell lines.
(A) MLPA; see Figure 1 for probe details. HeLa, MS751, SiHa, and HT3 (and HeLa subclone HeLaS3) contain distinctive homozygous deletions. C4I harbors a monoallelic deletion, as evidenced by contiguous probes with half-intensity signals. (B) Southern analysis of control DNA, above cell lines, C33 (no deletion by MLPA) and HPV16/E6E7-immortalized endocervical cells from a normal patient (Endo = End1/E6E7; ATCC #CRL-2615). (C) Western analysis. LKB1 protein is undetectable in lines harboring homozygous deletions and decreased by ∼50% in C4I, consistent with monoallelic loss.
Figure 4. LKB1 deletion breakpoints in cervical…
Figure 4. LKB1 deletion breakpoints in cervical cancer cell lines.
(A) LKB1 locus (chromosome 19p13.3). (B) 140 kb region (Ensembl50; 1050000–1190000) spanning LKB1 and immediately flanking loci; intervals = 10 kb. (C) Deletion breakpoints for cell lines harboring homozygous deletions. In MS751, deleted sequences are discontinuous as shown, consistent with a more complex rearrangement. Arrows = PCR primers for HeLa specific PCR (400 bp). (D) HeLa specific PCR (400 bp) confirms presence of deletion in HeLaS3. (E) HeLa intragenic LKB1 deletion occurred in vivo. Archival blocks were cored for DNA preparation. Lanes are as follows: (−) control = no template control; tumor = metastatic adrenal deposit; non-tumor = normal adrenal (same tissue block); (+) control = HeLa cell line DNA.
Figure 5. Progression-free survival of patients with…
Figure 5. Progression-free survival of patients with LKB1-wild type vs. LKB1-mutant tumors.
Kaplan-Meier curves show the percentage of patients with disease progression over time. The curves compare patients whose tumors were heterozygous or homozygous for mutations/deletions (LKB1) vs. patients with no mutations/deletions (wt). Patients with >1 follow-up visit were included in the analysis. P value per log-rank test.

References

    1. Schoell WM, Janicek MF, Mirhashemi R. Epidemiology and biology of cervical cancer. Semin Surg Oncol. 1999;16:203–211.
    1. Steben M, Duarte-Franco E. Human papillomavirus infection: epidemiology and pathophysiology. Gynecol Oncol. 2007;107:S2–5.
    1. Ellenson LH, Wu TC. Focus on endometrial and cervical cancer. Cancer Cell. 2004;5:533–538.
    1. Crum CP, Lee KR. Diagnostic Gynecologic and Obstetric Pathology. Carlsbad, CA: Saunders; 2006.
    1. Arbyn M, Bergeron C, Klinkhamer P, Martin-Hirsch P, Siebers AG, et al. Liquid compared with conventional cervical cytology: a systematic review and meta-analysis. Obstet Gynecol. 2008;111:167–177.
    1. Dall KL, Scarpini CG, Roberts I, Winder DM, Stanley MA, et al. Characterization of naturally occurring HPV16 integration sites isolated from cervical keratinocytes under noncompetitive conditions. Cancer Res. 2008;68:8249–8259.
    1. zur Hausen H. Condylomata acuminata and human genital cancer. Cancer Res. 1976;36:794.
    1. Alessi DR, Sakamoto K, Bayascas JR. Lkb1-dependent signaling pathways. Annu Rev Biochem. 2006;75:137–163.
    1. Ji H, Ramsey MR, Hayes DN, Fan C, McNamara K, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448:807–810.
    1. Matsumoto S, Iwakawa R, Takahashi K, Kohno T, Nakanishi Y, et al. Prevalence and specificity of LKB1 genetic alterations in lung cancers. Oncogene. 2007;26:5911–5918.
    1. Contreras CM, Gurumurthy S, Haynie JM, Shirley L, Akbay EA, et al. Loss of Lkb1 provokes highly invasive endometrial adenocarcinomas. Cancer Res. 2008;68:759–766.
    1. Gurumurthy S, Hezel AF, Berger JH, Bosenberg MW, Bardeesy N. LKB1 deficiency sensitizes mice to carcinogen-induced tumorigenesis. Cancer Res. 2008;68:55–63.
    1. Hearle N, Schumacher V, Menko FH, Olschwang S, Boardman LA, et al. Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res. 2006;12:3209–3215.
    1. Boudeau J, Kieloch A, Alessi DR, Stella A, Guanti G, et al. Functional analysis of LKB1/STK11 mutants and two aberrant isoforms found in Peutz-Jeghers Syndrome patients. Hum Mutat. 2003;21:172.
    1. Nezu J, Oku A, Shimane M. Loss of cytoplasmic retention ability of mutant LKB1 found in Peutz-Jeghers syndrome patients. Biochem Biophys Res Commun. 1999;261:750–755.
    1. Launonen V, Avizienyte E, Loukola A, Laiho P, Salovaara R, et al. No evidence of Peutz-Jeghers syndrome gene LKB1 involvement in left-sided colorectal carcinomas. Cancer Res. 2000;60:546–548.
    1. Bardeesy N, Sinha M, Hezel AF, Signoretti S, Hathaway NA, et al. Loss of the Lkb1 tumour suppressor provokes intestinal polyposis but resistance to transformation. Nature. 2002;419:162–167.
    1. Young RH, Clement PB. Endocervical adenocarcinoma and its variants: their morphology and differential diagnosis. Histopathology. 2002;41:185–207.
    1. Tiainen M, Ylikorkala A, Makela TP. Growth suppression by Lkb1 is mediated by a G(1) cell cycle arrest. Proc Natl Acad Sci U S A. 1999;96:9248–9251.
    1. Esteller M, Avizienyte E, Corn PG, Lothe RA, Baylin SB, et al. Epigenetic inactivation of LKB1 in primary tumors associated with the Peutz-Jeghers syndrome. Oncogene. 2000;19:164–168.
    1. Puck TT, Marcus PI. A Rapid Method for Viable Cell Titration and Clone Production with Hela Cells in Tissue Culture: The Use of X-Irradiated Cells to Supply Conditioning Factors. Proc Natl Acad Sci U S A. 1955;41:432–437.
    1. Gey GO, Coffman WD, Kubicek MT. Tissue culture studies of the proliferative capacity of cervical carcinoma and normal epithelium. Cancer Research. 1952;12:264–265.
    1. Connolly DC, Katabuchi H, Cliby WA, Cho KR. Somatic mutations in the STK11/LKB1 gene are uncommon in rare gynecological tumor types associated with Peutz-Jegher's syndrome. Am J Pathol. 2000;156:339–345.
    1. Avizienyte E, Loukola A, Roth S, Hemminki A, Tarkkanen M, et al. LKB1 somatic mutations in sporadic tumors. Am J Pathol. 1999;154:677–681.
    1. Masters JR. HeLa cells 50 years on: the good, the bad and the ugly. Nat Rev Cancer. 2002;2:315–319.
    1. Jones HW., Jr. Record of the first physician to see Henrietta Lacks at the Johns Hopkins Hospital: history of the beginning of the HeLa cell line. Am J Obstet Gynecol. 1997;176:S227–228.
    1. Jones HW, Jr., McKusick VA, Harper PS, Wuu KD. George Otto Gey. (1899–1970). The HeLa cell and a reappraisal of its origin. Obstet Gynecol. 1971;38:945–949.
    1. Hsu SH, Schacter BZ, Delaney NL, Miller TB, McKusick VA, et al. Genetic characteristics of the HeLa cell. Science. 1976;191:392–394.
    1. Pearson HB, McCarthy A, Collins CM, Ashworth A, Clarke AR. Lkb1 deficiency causes prostate neoplasia in the mouse. Cancer Res. 2008;68:2223–2232.
    1. Boshart M, Gissmann L, Ikenberg H, Kleinheinz A, Scheurlen W, et al. A new type of papillomavirus DNA, its presence in genital cancer biopsies and in cell lines derived from cervical cancer. EMBO J. 1984;3:1151–1157.
    1. Hezel AF, Bardeesy N. LKB1; linking cell structure and tumor suppression. Oncogene. 2008;27:6908–6919.
    1. Bonin S, Petrera F, Rosai J, Stanta G. DNA and RNA obtained from Bouin's fixed tissues. J Clin Pathol. 2005;58:313–316.

Source: PubMed

3
Suscribir