origins of lymphatic and distant metastases inhuman ... · tumor evolution origins of lymphatic and...

7
TUMOR EVOLUTION Origins of lymphatic and distant metastases in human colorectal cancer Kamila Naxerova, 1,2 * Johannes G. Reiter, 3 Elena Brachtel, 4 Jochen K. Lennerz, 4 Marc van de Wetering, 5,6 Andrew Rowan, 7 Tianxi Cai, 8 Hans Clevers, 5,6 Charles Swanton, 7,9 Martin A. Nowak, 3,10 Stephen J. Elledge, 2,11 Rakesh K. Jain 1 The spread of cancer cells from primary tumors to regional lymph nodes is often associated with reduced survival. One prevailing model to explain this association posits that fatal, distant metastases are seeded by lymph node metastases. This view provides a mechanistic basis for the TNM staging system and is the rationale for surgical resection of tumor-draining lymph nodes. Here we examine the evolutionary relationship between primary tumor, lymph node, and distant metastases in human colorectal cancer. Studying 213 archival biopsy samples from 17 patients, we used somatic variants in hypermutable DNA regions to reconstruct high-confidence phylogenetic trees. We found that in 65% of cases, lymphatic and distant metastases arose from independent subclones in the primary tumor, whereas in 35% of cases they shared common subclonal origin.Therefore, two different lineage relationships between lymphatic and distant metastases exist in colorectal cancer. T he spread of cancer cells from the primary tumor to regional lymph nodes is one of the most important factors predicting sur- vival in patients with epithelial cancers (1). Lymph node metastasis uniformly asso- ciates with worse outcomes in breast (2), pros- tate (3), lung (4), and colorectal cancer (5), the most frequent cancers in the U.S. population. In colorectal carcinoma, the presence of cancer cells in tumor-draining lymph nodes defines stage III disease and triggers administration of adjuvant chemotherapy (6). The 5-year survival for patients with stage II (no lymph node metastases) is 82.5%, in contrast to 59.5% for patients with stage III disease (7). In most patients, lymph node metastasis is not the cause of death but is correlated with spread to vital organs (8). The association between lym- phatic and distant metastasis has been known for at least 150 years (9) and, together with the observation that lymph node disease often pre- cedes systemic disease, has engendered the view that affected lymph nodes may give rise to dis- tant metastases (1012). The concept of such a sequential progression or metastatic cascade (13), in which the primary tumor (T) seeds lymph node metastases (N) that in turn seed distant metastases (M), provides a mechanistic basis for the TNM staging system. A corollary of the sequential-progression model is that surgical resection of positive lymph nodes will reduce recurrence rates. Indeed, resection of regional lymph nodes has been performed for more than 100 years (14). More recently, a number of clinical trials have shown that lymph node re- moval does not always improve patient survival (15). These findings have inspired the alterna- tive view that lymph node metastases do not give rise to distant metastases (16) and sug- gest that treatment strategies may need to be reevaluated (17). Given its potential impact on patient care, a better understanding of the evolutionary re- lationship between lymph node and distant metas- tases is critical. We still do not know whether a single metastatic subclone evolves in the primary tumor, subsequently spreading to lymph nodes and distant sites (1821), or whether multiple subclones in the primary tumor independently seed lymphatic and distant metastases ( 2224). Here we begin to examine these questions by studying the evolutionary history of colorectal cancer metastases. Insertion and deletion mutations (indels) in hypermutable DNA enable reconstruction of tumor phylogenies We conducted a systematic review of 1373 patient records and diagnostic materials at Massachu- setts General Hospital (MGH) (fig. S1) and ini- tially identified 19 colorectal cancer patients for whom formalin-fixed and paraffin-embedded samples from primary tumors, lymph nodes, and distant metastases were available (Fig. 1A). We collected multiple tumor regions for each patient (mean 12.6, range 7 to 29) for a total of 239 samples [92 primary tumor biopsies, 59 lymph node metastases, 52 distant metastases, 36 normal tissue (germline) samples] (table S1). Of the 19 patients, 17 had liver metastases, 1 had an ovary metastasis, and 1 had multiple metastases in the omentum. To trace the evolution of these cancers, we used a methodology (25) that leverages indel mutations in hypermutable, noncoding polygua- nine repeats (fig. S2). The mutation rate of poly- guanine repeats is several orders of magnitude greater than the mutation rate of nonrepetitive DNA (26), making these sequences a rich res- ervoir of neutral somatic variation. Previous work has shown that indels in polyguanine repeats (27), as well as other microsatellites (28), ac- curately reconstruct evolutionary events modeled in cell culture. Furthermore, in silico models of polyguanine-tract evolution have demonstrated that revertant or parallel mutations do not nota- bly affect phylogenetic reconstruction accuracy when the number of interrogated markers is larger than 10 (29). These properties make poly- guanine tracts attractive tools for phylogenetic analyses. Here, the mutation information from 20 to 43 polyguanine markers was generally suf- ficient to resolve lineages at our chosen confi- dence threshold (clade confidence > 70%, figs. S3 and S4). Polyguanine markers were distributed across many chromosomes (table S2). Therefore, any individual chromosomal alteration (gain or loss) would not be expected to substantially in- fluence phylogenetic reconstruction. In total, our data set consisted of 19,541 individual genotypes. We developed a fully automated pipeline for data filtering, noise reduction, and phylogenetic re- construction (supplementary methods). First, to avoid artifacts created by contamination with nor- mal cells, we implemented rigorous purity crite- ria, eliminating 11% of specimens from our study (fig. S5 and table S1). For all specimens belonging to the same patient, we then calculated a pairwise distance measure, the Jensen-Shannon distance (JSD) (30), over all polyguanine repeats. This distance reflected how much the samples had genetically diverged and formed the basis of our phylogenetic reconstruction with the neighbor- joining method (31). Indel mutation patterns in the colorectal can- cer cohort differed among patients. Of all observed alterations, 81% were deletions and 19% were insertions. A 4:1 ratio of deletions to insertions has previously been described by us and others (25, 32), indicating that it is an inherent property of polyguanine repeats. However, among indi- vidual cancers, we observed a relatively wide range of deletion frequencies, ranging from 45% in patient C69 to 100% in patient C77 (fig. S6). This suggests that additional determinants, such as alterations in specific DNA repair proteins, may contribute to a skewing of mutation patterns in individual tumors. For example, a cancer show- ing microsatellite instability due to loss of MLH1 protein expression almost exclusively harbored RESEARCH Naxerova et al., Science 357, 5560 (2017) 7 July 2017 1 of 6 1 Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. 2 Division of Genetics, Department of Genetics, Brigham and Womens Hospital and Harvard Medical School, Boston, MA 02115, USA. 3 Program for Evolutionary Dynamics, Harvard University, Cambridge, MA 02138, USA. 4 Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. 5 Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, 3584CT Utrecht, Netherlands. 6 Cancer Genomics Netherlands, UMC Utrecht, 3584CG Utrecht, Netherlands. 7 The Francis Crick Institute, London NW1 1AT, UK. 8 Department of Biostatistics, Harvard University, Boston, MA 02115, USA. 9 University College London Cancer Institute, London WC1E 6DD, UK. 10 Department of Mathematics and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA. 11 Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA. *Corresponding author. Email: [email protected] on January 11, 2021 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 21-Sep-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

TUMOR EVOLUTION

Origins of lymphatic and distantmetastases in human colorectal cancerKamila Naxerova,1,2* Johannes G. Reiter,3 Elena Brachtel,4 Jochen K. Lennerz,4

Marc van de Wetering,5,6 Andrew Rowan,7 Tianxi Cai,8 Hans Clevers,5,6

Charles Swanton,7,9 Martin A. Nowak,3,10 Stephen J. Elledge,2,11 Rakesh K. Jain1

The spread of cancer cells from primary tumors to regional lymph nodes is oftenassociated with reduced survival. One prevailing model to explain this association positsthat fatal, distant metastases are seeded by lymph node metastases. This view providesa mechanistic basis for the TNM staging system and is the rationale for surgical resectionof tumor-draining lymph nodes. Here we examine the evolutionary relationship betweenprimary tumor, lymph node, and distant metastases in human colorectal cancer. Studying213 archival biopsy samples from 17 patients, we used somatic variants in hypermutableDNA regions to reconstruct high-confidence phylogenetic trees. We found that in 65%of cases, lymphatic and distant metastases arose from independent subclones in theprimary tumor, whereas in 35% of cases they shared common subclonal origin. Therefore,two different lineage relationships between lymphatic and distant metastases exist incolorectal cancer.

The spread of cancer cells from the primarytumor to regional lymph nodes is one ofthe most important factors predicting sur-vival in patients with epithelial cancers (1).Lymph node metastasis uniformly asso-

ciates with worse outcomes in breast (2), pros-tate (3), lung (4), and colorectal cancer (5), themost frequent cancers in the U.S. population.In colorectal carcinoma, the presence of cancercells in tumor-draining lymph nodes definesstage III disease and triggers administration ofadjuvant chemotherapy (6). The 5-year survival forpatients with stage II (no lymph node metastases)is 82.5%, in contrast to 59.5% for patients withstage III disease (7).Inmost patients, lymph nodemetastasis is not

the cause of death but is correlated with spreadto vital organs (8). The association between lym-phatic and distant metastasis has been knownfor at least 150 years (9) and, together with theobservation that lymph node disease often pre-cedes systemic disease, has engendered the view

that affected lymph nodes may give rise to dis-tant metastases (10–12). The concept of such asequential progression ormetastatic cascade (13),in which the primary tumor (T) seeds lymphnode metastases (N) that in turn seed distantmetastases (M), provides a mechanistic basisfor the TNM staging system. A corollary of thesequential-progression model is that surgicalresection of positive lymph nodes will reducerecurrence rates. Indeed, resection of regionallymph nodes has been performed for morethan 100 years (14). More recently, a number ofclinical trials have shown that lymph node re-moval does not always improve patient survival(15). These findings have inspired the alterna-tive view that lymph node metastases do notgive rise to distant metastases (16) and sug-gest that treatment strategies may need to bereevaluated (17).Given its potential impact on patient care, a

better understanding of the evolutionary re-lationshipbetween lymphnodeanddistantmetas-tases is critical. We still do not know whether asinglemetastatic subclone evolves in the primarytumor, subsequently spreading to lymph nodesand distant sites (18–21), or whether multiplesubclones in the primary tumor independentlyseed lymphatic and distant metastases (22–24).Here we begin to examine these questions bystudying the evolutionary history of colorectalcancer metastases.

Insertion and deletion mutations(indels) in hypermutable DNA enablereconstruction of tumor phylogenies

We conducted a systematic review of 1373 patientrecords and diagnostic materials at Massachu-setts General Hospital (MGH) (fig. S1) and ini-tially identified 19 colorectal cancer patientsfor whom formalin-fixed and paraffin-embeddedsamples from primary tumors, lymph nodes,

and distant metastases were available (Fig. 1A).We collected multiple tumor regions for eachpatient (mean 12.6, range 7 to 29) for a total of239 samples [92 primary tumor biopsies, 59 lymphnodemetastases, 52 distantmetastases, 36 normaltissue (germline) samples] (table S1). Of the 19patients, 17 had liver metastases, 1 had an ovarymetastasis, and 1 had multiple metastases in theomentum.To trace the evolution of these cancers, we

used a methodology (25) that leverages indelmutations in hypermutable, noncoding polygua-nine repeats (fig. S2). The mutation rate of poly-guanine repeats is several orders of magnitudegreater than the mutation rate of nonrepetitiveDNA (26), making these sequences a rich res-ervoir of neutral somatic variation. Previousworkhas shown that indels in polyguanine repeats(27), as well as other microsatellites (28), ac-curately reconstruct evolutionary eventsmodeledin cell culture. Furthermore, in silico models ofpolyguanine-tract evolution have demonstratedthat revertant or parallel mutations do not nota-bly affect phylogenetic reconstruction accuracywhen the number of interrogated markers islarger than 10 (29). These properties make poly-guanine tracts attractive tools for phylogeneticanalyses. Here, the mutation information from20 to 43 polyguanine markers was generally suf-ficient to resolve lineages at our chosen confi-dence threshold (clade confidence > 70%, figs.S3 and S4). Polyguaninemarkerswere distributedacross many chromosomes (table S2). Therefore,any individual chromosomal alteration (gain orloss) would not be expected to substantially in-fluence phylogenetic reconstruction. In total, ourdata set consisted of 19,541 individual genotypes.We developed a fully automated pipeline for datafiltering, noise reduction, and phylogenetic re-construction (supplementary methods). First, toavoid artifacts created by contaminationwith nor-mal cells, we implemented rigorous purity crite-ria, eliminating 11% of specimens from our study(fig. S5 and table S1). For all specimens belongingto the same patient, we then calculated a pairwisedistance measure, the Jensen-Shannon distance(JSD) (30), over all polyguanine repeats. Thisdistance reflected how much the samples hadgenetically diverged and formed the basis of ourphylogenetic reconstruction with the neighbor-joining method (31).Indel mutation patterns in the colorectal can-

cer cohort differed among patients. Of all observedalterations, 81% were deletions and 19% wereinsertions. A 4:1 ratio of deletions to insertionshas previously been described by us and others(25, 32), indicating that it is an inherent propertyof polyguanine repeats. However, among indi-vidual cancers, we observed a relatively widerange of deletion frequencies, ranging from 45%in patient C69 to 100% in patient C77 (fig. S6).This suggests that additional determinants, suchas alterations in specific DNA repair proteins,may contribute to a skewing of mutation patternsin individual tumors. For example, a cancer show-ing microsatellite instability due to loss ofMLH1protein expression almost exclusively harbored

RESEARCH

Naxerova et al., Science 357, 55–60 (2017) 7 July 2017 1 of 6

1Edwin L. Steele Laboratories for Tumor Biology, Departmentof Radiation Oncology, Massachusetts General Hospital andHarvard Medical School, Boston, MA 02114, USA. 2Division ofGenetics, Department of Genetics, Brigham and Women’sHospital and Harvard Medical School, Boston, MA 02115,USA. 3Program for Evolutionary Dynamics, HarvardUniversity, Cambridge, MA 02138, USA. 4Department ofPathology, Massachusetts General Hospital and HarvardMedical School, Boston, MA 02114, USA. 5Hubrecht Institute,Royal Netherlands Academy of Arts and Sciences (KNAW)and University Medical Center (UMC) Utrecht, 3584CTUtrecht, Netherlands. 6Cancer Genomics Netherlands, UMCUtrecht, 3584CG Utrecht, Netherlands. 7The Francis CrickInstitute, London NW1 1AT, UK. 8Departmentof Biostatistics, Harvard University, Boston, MA 02115,USA. 9University College London Cancer Institute, LondonWC1E 6DD, UK. 10Department of Mathematics andDepartment of Organismic and Evolutionary Biology,Harvard University, Cambridge, MA 02138, USA. 11HowardHughes Medical Institute, Harvard Medical School, Boston,MA 02115, USA.*Corresponding author. Email: [email protected]

on January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 2: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

deletions (Fig. 1B) that also were of a considerablylarger size than deletions in microsatellite-stabletumors (Fig. 1C and fig. S6).Next, we explored whether accumulation of

polyguanine indels is a cancer-related processor whether these mutations can be found in

age-matched normal intestinal stem cells (ISCs).We analyzed DNA from 18 clonal expansionsof human ISCs. Stem cell donors for 12 of theseexpansions were children (ages 4 to 14), and6 expansions were from a 66-year-old adult(fig. S7). Adult ISCs had diverged significantly

farther from a polyclonal germline referencethan ISCs from children (Fig. 2A), suggestingthat polyguanine indels accumulate in normalISCs. We also observed a significant correla-tion between clonal mutation frequency and pa-tient age in our colorectal cancer cohort (fig. S8).

Naxerova et al., Science 357, 55–60 (2017) 7 July 2017 2 of 6

ACGGG...GGGAG

13x

100%

ACGGG...GGGAG

12x

ACGGG...GGGAG

11x

ACGGG...GGGAG

11x

100%

50%

50%

-0.65

-0.85

Nor

mal

Prim

ary

tum

orM

et

Satellite nodules(Sat1/2)

Primarytumor(P1-5)

Venousinvasion

(VI)

Omentalmetastases

(M1-3)

Lymphaticmetastasis

(L1)

e

llll

Lymphatic metastasis (L)

Distant metastasis (M)

Primary tumor (P)

Normal (N)

Sample collectionIndel detection across 20-43

hypermutable lociPhylogenetic

reconstruction

TACGGG...GGGAGA

13x

TACGGG...GGGAGA

12x

TACGGG...GGGAGA

12x

TACGGG...GGGAGA

12x

P LM

N

Primarytumor (P1-4)

Lymphaticmetastasis

(L1-10)

Livermeta-stasis

(M1a-c)

0.8

-0.8

N

M

P

L

N M P L

Calculation ofpairwise distances

Patient screeningLiver resections atMGH 2006-2013

n=1373

Colorectal cancer patients with available P, M, L, N

n=19

Fig. 1. Tracing tumor evolution through indels in hypermutable DNA.(A) Study design schematic. DNA samples from primary tumor (P),distant metastases (M), lymph node metastases (L), and normal tissue(germline) (N) from 19 colorectal cancer patients were genotyped across20 to 43 hypermutable polyguanine repeats. The genetic divergencebetween two samples is the average distance across all markers. Thenumber of consecutive guanines at a hypothetical locus is indicated inthe middle panel. Pairwise distances among all samples from a patientwere used as input for phylogenetic reconstruction with the neighbor-joining algorithm. (B) Anatomical sketch and raw data example for a

cancer (C38) with microsatellite instability. Mutations can be present invarying percentages of cells within a sample. Therefore, the distance of atumor sample to the normal reference is a continuous value. Theheatmap shows tumor-normal Jensen-Shannon distances across allsamples and polyguanine markers. Green, deletions (here, a negative signindicates deletions); purple, insertions. Note that the heatmap onlyshows a small part of the full data set for a patient. Pairwise distancesbetween all samples are used for phylogenetic reconstruction.(C) Anatomical sketch and raw data example for a microsatellite-stablecancer (C58). Heatmaps for all patients are provided in fig. S13.

RESEARCH | RESEARCH ARTICLEon January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 3: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

The mean clonal mutation frequency in nor-mal ISCs from the 66-year-old donor was lowerthan the mean frequency in cancers from age-matched (50- to 69-year-old) patients (Fig. 2B),but with considerable overlap of the two distribu-tions. The baseline polyguanine mutation bur-den of a colorectal cancer, therefore, partiallyconsists of alterations that are present in all in-testinal cells.

Two distinct patterns of metastaticdissemination exist in colorectal cancer

The main goal of our study was to illuminate theevolutionary relationship between lymphatic anddistant metastases. We aimed to sample lymph

nodes as comprehensively as possible and in-cluded 91.3% of resected positive nodes in ouranalysis (fig. S9, see supplementarymethods fora detailed description of lymph node inclusioncriteria). We first investigated the genetic distancesamong lymph node metastases, primary tumorbiopsies, and distant metastases. For 33 of 45(73%) lymph nodemetastases, the distance to theprimary tumor (Fig. 2C) was shorter than the dis-tance to distant metastases, and 31 of 45 (69%)distantmetastases had a shorter genetic distanceto the primary tumor than to any lymph nodemetastasis (Fig. 2D). This indicates that bothtypes of metastatic lesions likely originated fromdistinct subclones in the primary tumor inmost

cases. To test this hypothesis, we examined allphylogenetic trees according to formal criteria.Patients were classified into two categories onthe basis of tree topology (Fig. 2E). We reasonedthat lymph node and distant metastases had acommon origin if a patient’s tree contained aclade that included at least one lymph node andat least one distant metastasis but no primarytumor samples. Existence of such a branch indi-cates that both types of metastases were seededfrom the same subclone, or that lymphnodemetas-tases gave rise to distant metastases. Formally, thereverse—seeding of lymphatic metastases fromdistant metastases—is also possible. A patientwas classified as having distinct origins of lym-phatic and distant metastases if no such cladeexisted. In all distinct origin cases, lymphatic anddistant metastases were eachmore closely relatedto a primary tumor region than to each other.To assess the robustness of each tumor’s origin

classification, we employed a bootstrapping strat-egy. We performed repeated random sampling(n = 1000) of a tumor’s mutation data to deter-minewhether our origin classificationwas sensitiveto changes in a limited number of polyguaninemarkers (supplementary methods). Of the 17 tu-mors sampled, 14 (82%) were classified with abootstrap value above 80% (Fig. 2F), confirmingthe robustness of our phylogenetic data and clas-sification scheme. We also evaluated our clas-sification by utilizing the unweighted pair-groupmethodwith arithmeticmean (UPGMA) (33). (Thenature of polyguanine genotyping data suggeststhe use of distance-based phylogenetic methods;see supplementarymethods). Origin classificationoutcomes did not change for any of our 17 patients,further demonstrating the reliability of our results(all phylogenetic trees are available at Dryad:http://dx.doi.org/10.5061/dryad.vv53d).

Common origin of lymphatic anddistant metastases

In 6 out of 17 tumors (35%), we found a commonorigin of lymphatic and distant metastases.Selected phylogenetic trees with a classificationconfidence score above 80% for common originare displayed in Fig. 3, along with pertinentclinical information.Patient C38’s cancer (Fig. 3A and anatomical

sketch in Fig. 1B) spread to the omentum and tothe mesenteric lymph nodes. Furthermore, sev-eral satellite nodules had formed within the co-lonic epithelium, spatially separated from theprimary tumor. We also investigated a piece oftumor that had invaded a vein. Notably, phylo-genetic reconstruction showed that all lesionswhose formation had depended on cell migra-tion (that is, the satellite nodules, the distantand lymph nodemetastases, and the tumor with-in the vein) shared common ancestry, where-as the primary tumor had a divergent geneticprofile.Patient C69’s cancer (Fig. 3B) showed a similar

pattern, with one metastatic subclone giving riseto several liver metastases and a lymph nodemetastasis. Polyguanine indels clearly attributedall metastases to the same evolutionary branch,

Naxerova et al., Science 357, 55–60 (2017) 7 July 2017 3 of 6

0.0

0.2

0.4

0.02

0.04

0.0

0.5

1.0

0.0

0.5

1.0

1.5

0.5

1.0

0.5

1.0

Children Adult

Dis

tanc

e to

ger

mlin

eD

ista

nce

ratio

Dis

tanc

e ra

tio

30-49 50-69 70-90 NormalISCs (66)

Clo

nal m

ut. f

requ

ency

p=0.001

L closer to PL closer to M

M closer to PM closer to L

L

NP1

M

P2

LN

M

P

Common origin (C)

Distinct origins (D)

C45C63C31C59C46C36C61C69C38C66C77C12C65C58C51C39C530

20

40

60

80

100

Boo

tstr

ap c

onfid

ence

(%

)

CD

Fig. 2. Common versus distinct origins of lymph node and distant metastases. (A) Clonalexpansions of single ISCs from children (age < 15 years old, n = 12) have fewer polyguanine indelsthan clonal expansions from a 66-year-old adult (n = 6). Data are mean ± SEM, two-tailedStudent’s t test. (B) Clonal mutation frequency in cancers (defined as JSD ≥ 0.11 in 95% of tumorbiopsies) is correlated with patient age at diagnosis. Normal ISCs, on average, have fewerpolyguanine indels than age-matched cancers, but the two distributions overlap. Lines indicate themean. (C) Most lymph node metastases are more closely related to the primary tumor than todistant metastases. The plot shows d(L to M)/d(L to P) – 1, the distance of each lymph node metastasis(L) to its closest distant metastasis (M), divided by its distance to its closest primary tumorsample (P), minus one. Yellow, closest neighbor is a metastasis; dark blue, closest neighbor is aprimary tumor sample. (D) Analogous plot for distant metastases, showing d(M to L)/d(M to P) – 1.(E) Classification of patients into cases with common or distinct origins of lymphatic and distantmetastases. (F) Bootstrap values reflecting origin-classification confidence for each patient(bootstrap n = 1000).

RESEARCH | RESEARCH ARTICLEon January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 4: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

even though M1, M2, and L1 were resected sev-eral months earlier than M3 and the patient re-ceived chemotherapy and bevacizumab betweensurgeries (M and L indicate distant metastasesand lymph node metastases, respectively).Patient C58 (Fig. 3C and the anatomical sketch

in Fig. 1C) had widespreadmetastases to the mes-enteric lymph nodes and the liver. Most lymphnodemetastaseswere closely related to theprimarytumor. However, a distinct subclone had formedin several lymph nodes that were located in closeanatomical proximity (L3, L5, L6). The liver me-tastasis derived from the same subclone found inthis lymph node group.Further examples of patients with a common

origin of lymphatic and distant metastases areshown in Fig. 3D and fig. S3. In all commonorigin cases, tree topologies are consistent withone of two modes of dissemination: The primarytumor seeded lymph node metastases, which inturn seeded distant metastases, with the formalpossibility of the reverse; or one genetically dis-tinct ancestor evolved within the primary tumorand subsequently colonized lymph nodes anddistant sites. In both scenarios, lymphatic anddistant metastases share a common origin.The common origin category is compatible

with the idea of sequential progression and can

explain important clinical observations, such as thewell-established correlation between lymphaticand distant disease. In amajority of patients, how-ever, tree topologies indicated independent seed-ing of lymphatic and distant metastasis from theprimary tumor.

Distinct origins of lymphatic anddistant metastases

Cancers in the distinct origins group, which en-compassed 11 out of 17 patients (65%), containedmultiple, genetically distinct metastasis ancestors.Figure 4 shows selected phylogenetic trees witha distinct origin–classification confidence scoreabove 80% (the complete set, along with confi-dence values for each clade, is provided in fig. S4).Patient C66’s tumor (Fig. 4A) is a represen-

tative example of the distinct origins category.The cancer harbored multiple subclones at dif-ferent stages of evolution that had seeded genet-ically distinct metastases. Area P2, for example,was most closely related to lymph node metas-tasis L3, whereas area P1 was the origin of livermetastases M1 and M2 (P indicates primary tu-mor). The tree shows that lymph node metastaseswere seeded continuously throughout the devel-opment of the tumor but did not metastasizefurther. Conversely, the liver metastases arose in

later evolution stages from the genetically mostadvanced clone. They constitute the terminal,most mutation-rich branch of the tree and, asa group, are more homogeneous than the lymphnode metastases.Patient C12’s tumor (Fig. 4B) partially resem-

bled that of patient C58. Its phylogenetic treealso showed a group of lymph node metastases(L2, L3, L4) that either derived from the sameancestral clone or gave rise to each other, where-as other lymphatic lesions (L1) were seeded inde-pendently. Notably, as for patient C58, the closelyrelated nodes also were in anatomical proximity.However, the patient’s liver metastasis (M1) didnot arise from this subclone but instead haddistinct origins in primary tumor area P8.Another noteworthy case from the distinct ori-

gins group is patient C53 (Fig. 4C), whounderwentresection of two metastases located in the rightliver lobe and one metastasis in the left liverlobe. Phylogenetic reconstruction showed that themetastases in the right liver diverged relativelyearly. After their divergence, the primary tumorevolved further and independently gave rise tolymph node metastasis L1 and the left liver me-tastasis M3.Further examples of cancers in the distinct

origins category are shown in Fig. 4, D to F,

Naxerova et al., Science 357, 55–60 (2017) 7 July 2017 4 of 6

M1a

M1b

L1

L2

P2

P1

N1

N2

C77

C38

P5P3P2

P1

P4

M2M1

M3

VISat1

Sat2

L1

N1

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:A

LIV

E

2.4 mo 56.4 mo

No

ther

apy

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (1/1)

Synchr.Complete

No

N1

M1b

L1

P1

N2

N3

P2

P3

P4

M1a

M1c

L2

L4

L5L6

L7L8

L10

L9

C58D

iagn

osis

Res

ectio

n (

P, L

, M)

Last

FU

:A

LIV

E

0.4 mo 36.9 mo

No

ther

apy

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 67% (10/15)

Synchr.Complete

No

C69

N1

N2 M3c M3b

M3aM2

M1

L1

P6

P5

P3P2P1

P4

Dia

gnos

is

Res

ectio

n (

P, L

, M1/

2)

Last

FU

: A

LIV

E

1.2 mo

Rad

iatio

n

Fol

fox/

Bev

Res

ectio

n (

M3)

6.0 mo 60.7 mo

SO

CS

OC

SO

C

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:D

OD

5.7 mo 45.7 mo

Rad

iatio

nF

olfo

x/B

ev

SO

C

Class. confidence 90.7%

99.5%Class. confidence

L3

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (2/2)

Synchr.Complete

Chemo/Rad.

Class. confidence 96.4%

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (1/1)

Syn/metaCompleteRadiation

Class. confidence 86.6%

Fig. 3. Phylogenetic trees of cancers with a common origin of lym-phatic and distant metastases. (A to D) All trees except C38 [micro-satellite-instability (MSI) case] (A) are drawn to scale and were constructedwith the neighbor-joining method. Seeding events [internal node (commonancestor) and branches] that gave rise to distant metastases are shadedin red; events that gave rise to lymph node metastases are shaded inblue. Clinical information boxes show whether a patient received neoadjuvanttherapy, whether primary tumor resection was complete (all margins

unaffected), whether distant metastases occurred synchronously or metach-ronously, what percentage of suitable lymph nodes (i.e., those thatwere large and pure enough, see supplementary methods for details) wassampled, and the origin classification bootstrap value. Timelines summarizetreatment and known life span for each patient. Lowercase letters (a, b, c)after sample numbers indicate multiple biopsies from the same tumormass. VI, venous invasion; Sat, satellite nodule; SOC, standard of care; FU,follow up; DOD, dead of disease.

RESEARCH | RESEARCH ARTICLEon January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 5: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

and fig. S4. In all these cases, the phylogeneticdata indicate that lymph node metastases werenot the source of distant metastases (also see ex-planatory schematic in fig. S10).

Common clinicopathologicalvariables do not correlate withorigin classification

Next, we examined whether our origin classi-fication was correlated with (and thus poten-tially influenced by) any clinicopathologicalvariables. We did not observe any significantdifferences in the number of positive nodes,the ratio of positive to examined nodes, thenumber of lymph nodes included in the finaldata set, the number of excluded nodes (fig.S11, A to D), the number of sampled primarytumor regions, the percentage of T3 versus T4stage patients (no T1 or T2 stage tumors werepart of this cohort), the distribution of primarytumor sizes, the presence of vascular invasion,or the fraction of patients with synchronous ver-sus metachronous distant metastasis (fig. S12, Ato E) between origin categories.

Most importantly, we found no associationbetween origin and treatment history. Onlyone patient (C77) had neoadjuvant chemother-apy (table S3). In six patients, distant metastaseswere resected after the primary tumor and thelymph node metastases had already been re-moved, and all received treatment in the inter-vening time interval. Three of these patients(C69, C65, C36) fell into the common origins andthree (C66, C39, C63) into the distinct originscategory (P = 0.6).

Discussion

The presence of lymph node metastases is animportant prognostic factor for most cancers,but the underlying reason has been unclear.One prevailing model posits that lymph nodemetastases are precursors of distant metastases,and their surgical resection is necessary to attaina “cancer-free” state (34). An alternative modelposits that distant metastases arise independentlyof lymph node metastases (16).Our data show that lymph node metastases

and distant metastases indeed often do have a

common origin. Although our phylogenies donot allow us to distinguish between sequentialprogression and common-subclonal origin, manyphylogenies in the common origin category arecompatible with seeding of distant metastasesfrom lymph nodes.However, in a majority of patients, we find

strong evidence of independent origins of lymphnode and distant lesions. If independent seedingis prevalent, what is the reason for the associa-tion of lymphatic and distant metastasis? It couldbe that the association is driven by the commonorigin subset of patients. An alternative possibilityis that most cells in tumors belonging to the dis-tinct origins category have the ability to metas-tasize. In such tumors, all cells that disseminatewould have an increased likelihood of colonizingdistant sites (35). Establishing lymph node metas-tases may be a more efficient process than es-tablishing distant metastases and may thereforehappen earlier and more frequently. This modelwould also be compatible with clinical obser-vations, including the correlation between lym-phatic and distant metastasis, the sometimes

Naxerova et al., Science 357, 55–60 (2017) 7 July 2017 5 of 6

P5

N1N2

P1P2

P3

P4M1

L1

M1a

M1b

L1

L2

P1

P2

P3

N1N2

N1 M2r

P1

P2P3

P4L1

M1br M1ar

M3l

L2L3

L4

L1

P1

P2

P3

P4

P6

P9

P8

P10P7

P5

M1

N1N2

N1N2

L1

L2

L3

L4

L5

P1P2 P3

M1bM1a

M2

C66 C12

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:D

OD

0.67 mo 2.2 mo

No

ther

apy

SO

C

C53

Dia

gnos

is

Res

ectio

n (

P, L

)

Last

FU

: D

OD

unknown

Fol

fox/

Bev

Res

ectio

n (

M1,

2)

9.1 mo 40.97 mo

SO

C

No

ther

apy

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:A

LIV

E

0.67 mo 44.37 mo

No

ther

apy

SO

C

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (1/1)

Synchr.Complete

No

Class. confidence

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (5/5)

Metachr.Complete

No

Class. confidence 96.3%

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 80% (4/5)

Synchr.Incomplete

No

Class. confidence 97.8%

100%

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:D

OD

unknown 24.97 mo

No

ther

apy

SO

C

C46

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (1/1)

Synchr.Complete

No

Class. confidence 80.6%

M1aM1b M1c

L1L2

L3

P1

P2

P3

P5

P4

N1

N2

C59

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:A

LIV

E

0.5 mo 42.6 mo

No

ther

apy

SO

C

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (3/3)

Synchr.Complete

No

Class. confidence 80.5%

C51

Dia

gnos

is

Res

ectio

n (

P, L

, M)

Last

FU

:D

OD

unknown 11 mo

No

ther

apy

SO

C

Primary resectionNeoadj. therapy

Metastasis timing% of LN sampled 100% (2/2)

Synchr.Complete

No

Class. confidence 99.5%

Fig. 4. Phylogenetic trees of cancers with distinct origins of lymphatic and distant metastases. (A to F) All trees except C12 (MSI case) (B) aredrawn to scale and were constructed with the neighbor-joining method. Shading and clinical information is as noted for Fig. 3. r, right liver; l, left liver.

RESEARCH | RESEARCH ARTICLEon January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 6: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

modest benefits of lymphadenectomy, and theadvantage of early primary resection (assumingthat even in such highly metastatic cancers, thesurvival rate of disseminated cells is relatively low,so that the tumor needs to grow to a certain sizein order to metastasize efficiently).Most metastases in our cohort were resected

from the liver, which is themost frequent distantsite of colorectal metastasis (36). Because venousblood from the intestines reaches the liver di-rectly through the portal vein, it is possible thatliver metastases are preferentially seeded hema-togenously. Cancer cells that migrate throughlymph nodes enter the venous circulation in thesubclavian vein. The first capillary bed that suchcells encounter is the lung. It is therefore possiblethat lung metastases are more frequently seededthrough the lymph nodes.All cancers in our study were retrospectively

collected specimens. Archival samples aremostlynot suitable for whole-genome or exome sequenc-ing because patient consent for such comprehen-sive genetic profiling was not obtained at thetime of surgery. Conversely, polyguanine-repeatgenotyping is a limited analysis of length poly-morphisms in noncodingDNA. It does not produceany information about functional or disease-related genes. Raw data produced by ourmethodcontain the lengths of polymerase chain reaction(PCR) amplicons in arbitrary units, allowing forcomplete disclosure ofmutation informationwhilemaking patient identification impossible. There-fore, polyguanine-repeat analysis represents asafe and effective method for studying tumorevolution in a patient population that wouldotherwise be inaccessible.We conclude that the evolutionary relation-

ship between lymphatic and distant metastasescan take on two different forms in colorectalcancer. In the future, it will be important to de-

termine whether cancers in the common anddistinct origin categories exhibit different clin-ical behaviors.

REFERENCES AND NOTES

1. S. D. Nathanson, Cancer 98, 413–423 (2003).2. W. L. McGuire, Breast Cancer Res. Treat. 10, 5–9 (1987).3. L. A. Gervasi et al., J. Urol. 142, 332–336 (1989).4. T. Naruke, K. Suemasu, S. Ishikawa, J. Thorac. Cardiovasc.

Surg. 76, 832–839 (1978).5. G. J. Chang, M. A. Rodriguez-Bigas, J. M. Skibber, V. A. Moyer,

J. Natl. Cancer Inst. 99, 433–441 (2007).6. American Cancer Society, Treatment of colon cancer, by stage;

www.cancer.org/cancer/colon-rectal-cancer/treating/by-stage-colon.html.

7. J. B. O’Connell, M. A. Maggard, C. Y. Ko, J. Natl. Cancer Inst.96, 1420–1425 (2004).

8. S. Y. Wong, R. O. Hynes, Cell Cycle 5, 812–817 (2006).9. R. Virchow, Die krankhaften Geschwülste: Dreissig Vorlesungen

gehalten während des Wintersemesters 1862–1893 an derUniversität zu Berlin (Verlag von August Hirschwald, Berlin,1863).

10. C. M. McBride, South. Med. J. 71, 1331–1333 (1978).11. W. S. Halsted, Ann. Surg. 46, 1–19 (1907).12. J. Sleeman, A. Schmid, W. Thiele, Semin. Cancer Biol. 19,

285–297 (2009).13. R. A. Weinberg, Carcinogenesis 29, 1092–1095 (2008).14. B. Moynihan, Surg. Gynecol. Obstet. 6, 463–466 (1908).15. J. E. Gervasoni Jr., S. Sbayi, B. Cady, Ann. Surg. Oncol. 14,

2443–2462 (2007).16. B. Cady, Arch. Surg. 119, 1067–1072 (1984).17. J. Engel, R. T. Emeny, D. Hölzel, Cancer Metastasis Rev. 31,

235–246 (2012).18. W. Liu et al., Nat. Med. 15, 559–565 (2009).19. A. McPherson et al., Nat. Genet. 48, 758–767 (2016).20. W. J. Gibson et al., Nat. Genet. 48, 848–855 (2016).21. M. Gerlinger et al., N. Engl. J. Med. 366, 883–892

(2012).22. S. Yachida et al., Nature 467, 1114–1117 (2010).23. J. G. Reiter et al., Nat. Commun. 8, 14114 (2017).24. M. C. Haffner et al., J. Clin. Invest. 123, 4918–4922

(2013).25. K. Naxerova et al., Proc. Natl. Acad. Sci. U.S.A. 111,

E1889–E1898 (2014).26. J. C. Boyer et al., Hum. Mol. Genet. 11, 707–713 (2002).27. S. J. Salipante, M. S. Horwitz, Proc. Natl. Acad. Sci. U.S.A. 103,

5448–5453 (2006).28. D. Frumkin, A. Wasserstrom, S. Kaplan, U. Feige, E. Shapiro,

PLOS Comput. Biol. 1, e50 (2005).

29. S. J. Salipante, J. M. Thompson, M. S. Horwitz, Genetics 178,967–977 (2008).

30. D. M. Endres, J. E. Schindelin, IEEE Trans. Inf. Theory 49,1858–1860 (2003).

31. N. Saitou, M. Nei, Mol. Biol. Evol. 4, 406–425 (1987).32. J. J. Salk et al., Proc. Natl. Acad. Sci. U.S.A. 106, 20871–20876

(2009).33. R. R. Sokal, Univ. Kans. Sci. Bull. 38, 1409–1438 (1958).34. J. P. Sleeman, B. Cady, K. Pantel, Clin. Exp. Metastasis 29,

737–746 (2012).35. B. Vogelstein, K. W. Kinzler, N. Engl. J. Med. 373, 1895–1898

(2015).36. V. Patanaphan, O. M. Salazar, South. Med. J. 86, 38–41

(1993).

ACKNOWLEDGMENTS

We thank M. Nahrendorf, F. Swirski, J. Gerold, and T. Padera forhelpful comments and careful review of the manuscript, andN. Sasaki and V. Sasselli for their help with organoid culture. Thiswork was supported by the Department of Defense W81XWH-10-0016 (R.K.J.), W81XWH-12-1-0362 (S.J.E.), and W81XWH-15-1-0579(K.N.); National Human Genome Research Institute U54 HG007963(T.C.); National Cancer Institute P01-CA080124 (R.K.J.) andR35-CA197743 (R.K.J.); Francis Crick Institute FC001169 (C.S.);Austrian Science Fund J-3996 (J.G.R.); National Foundation forCancer Research (R.K.J.); and Ludwig Center at Harvard (S.J.E.and R.K.J.). The Program for Evolutionary Dynamics is supported inpart by a gift from B. Wu and E. Larson. K.N. conceived anddesigned the study and performed experiments. K.N. and J.G.R.analyzed data. E.B. and J.K.L. reviewed tissue specimens andclinical records. M.v.d.W., A.R., H.C., and C.S. provided DNAsamples. K.N., J.G.R., E.B., J.K.L., T.C., C.S., M.A.N., S.J.E., andR.K.J. discussed results and strategy. R.K.J. supervised the study.K.N. wrote the manuscript, which was revised and approved byall authors. Raw polyguanine-profiling data, distance matrices, andphylogenetic trees can be downloaded from https://steelelabs.mgh.harvard.edu/lymphmet and from datadryad.org (http://dx.doi.org/10.5061/dryad.vv53d).

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/357/6346/55/suppl/DC1Materials and MethodsFigs. S1 to S14Tables S1 to S3References

23 August 2016; resubmitted 16 February 2017Accepted 8 May 201710.1126/science.aai8515

Naxerova et al., Science 357, 55–60 (2017) 7 July 2017 6 of 6

RESEARCH | RESEARCH ARTICLEon January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from

Page 7: Origins of lymphatic and distant metastases inhuman ... · TUMOR EVOLUTION Origins of lymphatic and distant metastases inhuman colorectal cancer Kamila Naxerova,1,2* Johannes G. Reiter,3

Origins of lymphatic and distant metastases in human colorectal cancer

Hans Clevers, Charles Swanton, Martin A. Nowak, Stephen J. Elledge and Rakesh K. JainKamila Naxerova, Johannes G. Reiter, Elena Brachtel, Jochen K. Lennerz, Marc van de Wetering, Andrew Rowan, Tianxi Cai,

DOI: 10.1126/science.aai8515 (6346), 55-60.357Science 

, this issue p. 55; see also p. 35Sciencewithin the primary tumor.patients. In the other two-thirds, distant metastases and lymph node metastases originated from independent subclones colorectal cancer (see the Perspective by Markowitz). The sequential progression model applied to only one-third of thethe evolutionary relationship of primary tumors, lymph node metastases, and distant metastases in 17 patients with

used phylogenetic methods to reconstructet al.rationale for surgical removal of tumor-draining lymph nodes. Naxerova in oncology is that lymph node metastases give rise to distant metastases. This ''sequential progression model'' is the

Cancer cells from primary tumors can migrate to regional lymph nodes and distant organs. The prevailing modelMetastases undergo reconstruction

ARTICLE TOOLS http://science.sciencemag.org/content/357/6346/55

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2017/07/06/357.6346.55.DC1

CONTENTRELATED

http://stm.sciencemag.org/content/scitransmed/9/384/eaai8504.fullhttp://stm.sciencemag.org/content/scitransmed/9/397/eaan0026.fullhttp://stm.sciencemag.org/content/scitransmed/8/361/361ra138.fullhttp://stm.sciencemag.org/content/scitransmed/8/364/364ra153.full

REFERENCES

http://science.sciencemag.org/content/357/6346/55#BIBLThis article cites 34 articles, 4 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Science. No claim to original U.S. Government WorksCopyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on January 11, 2021

http://science.sciencemag.org/

Dow

nloaded from