Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2015 Oct 12;112(43):13308–13311. doi: 10.1073/pnas.1516689112

Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases

Fleur Weeber a, Marc van de Wetering b,c,d, Marlous Hoogstraat e, Krijn K Dijkstra a, Oscar Krijgsman a, Thomas Kuilman a, Christa G M Gadellaa-van Hooijdonk f, Daphne L van der Velden a, Daniel S Peeper a, Edwin P J G Cuppen b,c,g, Robert G Vries b,c,d, Hans Clevers b,c,d,h,1, Emile E Voest a,c,d,i,1
PMCID: PMC4629330  PMID: 26460009

Significance

Chemotherapy has been proven in clinical studies to improve overall survival significantly. Unfortunately, there is a significant degree of heterogeneity in tumor chemosensitivity, often resulting in unnecessary treatment and needless exposure to toxic side-effects. A platform is needed that can identify preemptively which patients will or will not benefit from treatment. Tumor organoids, 3D cultures of cancer cells, present such an individualized platform. In this study we demonstrate that organoid cultures can be established from metastatic biopsy specimens with a high success rate and genetically represent the metastasis they were derived from. These data support the translation of this innovative technology to the clinic as an ex vivo screening platform for tailoring treatment.

Keywords: biopsy-derived tumor organoids, colorectal cancer, DNA sequencing, copy number analysis, personalized medicine

Abstract

Tumor organoids are 3D cultures of cancer cells. They can be derived from the tumor of each individual patient, thereby providing an attractive ex vivo assay to tailor treatment. Using patient-derived tumor organoids for this purpose requires that organoids derived from biopsies maintain the genetic diversity of the in vivo tumor. In this study tumor biopsies were obtained from 14 patients with metastatic colorectal cancer (i) to test the feasibility of organoid culture from metastatic biopsy specimens and (ii) to compare the genetic diversity of patient-derived tumor organoids and the original tumor biopsy. Genetic analysis was performed using SOLiD sequencing for 1,977 cancer-relevant genes. Copy number profiles were generated from sequencing data using CopywriteR. Here we demonstrate that organoid cultures can be established from tumor biopsies of patients with metastatic colorectal cancer with a success rate of 71%. Genetic analysis showed that organoids reflect the metastasis from which they were derived. Ninety percent of somatic mutations were shared between organoids and biopsies from the same patient, and the DNA copy number profiles of organoids and the corresponding original tumor show a correlation of 0.89. Most importantly, none of the mutations that were found exclusively in either the tumor or organoid culture are in driver genes or genes amenable for drug targeting. These findings support further exploration of patient-derived organoids as an ex vivo platform to personalize anticancer treatment.


The considerable variation among patients in sensitivity to antineoplastic treatment emphasizes the need for more accurate treatment selection. Numerous attempts have been made to develop a personalized in vitro platform to predict treatment response in individual patients, but no method has presently gained clinical acceptance (1). Cell lines are not readily established for every individual patient, and although a higher take rate and early signs of predictive value have been reported for patient-derived xenograft models, the 6–8 mo required to gain sufficient material may prohibit its development as a clinical decision-making tool (2).

A recently developed 3D culture system, enriching for the stem cell population, has enabled the indefinite propagation of normal and tumor epithelial cells of a variety of tissues (39). These cultures, referred to as “organoids,” are established in a relatively short time frame, are easy to manipulate, and facilitate high-throughput screens (10). However, their most important feature is that cultures can be derived from the tumor tissue of each individual patient. With adequate amounts of starting material, such as tumor resections, the success rate of derivation for colorectal cancer organoids is greater than 90% (10). However, in the metastatic setting there generally is no access to resection material, and acquisition of fresh tumor tissue often is limited to biopsies. To use patient-derived tumor organoids as a personalized screening tool for tailoring treatment, it is imperative that cultures from biopsy specimens have a high success rate and that the genetic landscape of the tumor be preserved in culture. This article addresses both topics.

Materials and Methods

Patients.

Fourteen patients with metastatic colorectal cancer gave written informed consent to undergo two to four 18-gauge biopsies from accessible metastatic lesions (Fig. 1). This study was approved by the institutional review board of the University Medical Center Utrecht (protocol number NCT01855477).

Fig. 1.

Fig. 1.

Overview of the study design.

Organoid Cultures.

Organoid culturing was performed according to methods described by Sato et al. (3). Briefly, biopsies were dissected and embedded in Matrigel [Basement Membrane Matrix, growth Factor Reduced, Phenol Red free (MG), (BD; #56231)]. The Matrigel including the embedded cells was overlaid with growth medium optimized for selective outgrowth of tumor cells [Advanced Dulbecco's Modified Eagles Medium with Nutrient Mixture F-12 Hams (Ad-DF) (Invitrogen; #12634), 1% penicillin/streptomycin (Invitrogen; #15140-122), 10 mM Hepes (Invitrogen #15630-056), 1% GlutaMAX (Invitrogen #35050), 20% R-Spondin conditioned medium (3), 10% Noggin conditioned medium (3), 1× B27 supplement (Invitrogen #17504-044), 1.25 mM N-acetyl-cysteine (Sigma-Aldrich; A9165-5G), 10 mM nicotinamide (Sigma-Aldrich; N0636), 50 ng/mL human EGF (Invitrogen Biosource; PMG8043), 10 nM gastrin (Sigma; #G9145), 500 nM TGFb type I Receptor inhibitor A83-01 (Tocris; #2939), 10 μM p38 MAPK inhibitor (p38i) SB202190 (Sigma-Aldrich; S7067), 10 nM prostaglandin E2 (Tocris; #2296), and 1× Primocin (InvivoGen; #ant-pm-1)]. Fresh medium was added every 2 or 3 d. Outgrowing organoids were passaged every 7–10 d after mechanical and enzymatic disruption.

DNA Sequencing.

A separate biopsy was used to compare the mutation and copy number profiles of the tumor biopsy and biopsy-derived cultures. Blood was drawn to obtain control DNA for germline variation. DNA was isolated from organoid cultures after 2–3 mo (depending on organoid growth rate). DNA was isolated using the Tissue and Blood DNA 500 Extraction Kit (Diasorin) according to the manufacturer’s protocol. Matched blood, tumor biopsies, and biopsy-derived cultures were sequenced using the 5500xl SOLiD system (Applied Biosystems) according to the manufacturer’s protocol. The minimum tumor percentage required to enable sequencing was 30%. Tumor cell percentage and percentage of necrosis were determined by a trained pathologist based on H&E staining. Six hundred nanograms of genomic DNA were required per sample to generate barcoded fragment libraries as described by Harakalova et al. (11). Samples were enriched using SureSelect technology (Agilent Technologies) for the actionable cancer genome consisting of 1,977 genes, as previously described by Hoogstraat et al. (12) and Vermaat et al. (13). The actionable cancer genome consists of all exons of genes known to be oncogenes, tumor-suppressor genes, kinases, or functioning in pathways involved in tumorigenesis and comprises ∼16% of the exome. Libraries were sequenced to an average coverage of 150×. Mapping, variant calling, and annotation were done as described by Hoogstraat et al. (12). Somatic mutations were classified as unique for either tumor biopsy or biopsy-derived cultures if the allele frequency in the matching sample was <1%.

DNA Copy Number Analysis.

DNA copy number profiles were generated from the BAM files using CopywriteR as described by Kuilman et al. (14). In short, sequence reads outside the captured genomic regions (off-target reads) were used to generate DNA copy number profiles. A depth-of-coverage method was used for 100-kb bins, and the read count was normalized for GC content and mappability. Log2 ratios were calculated for all tumor or organoid samples versus reference (blood) samples. The normalized and corrected log2 ratios from CopywriteR were analyzed further by circular binary segmentation (CBS) and CGHcall (Bioconductor) (15). CBS allows the detection of segments with nearly identical copy number states. CGHcall is used to classify data points as copy number gain, loss, or neutral.

Results

Organoid culture from metastatic biopsy specimens was successful in 10 of 14 cases (71%). For eight patients DNA from the tumor biopsy and its matched biopsy-derived culture were sequenced to assess whether the original genetic diversity of the tumor was preserved in culture (Fig. 1). The remaining two patients from whom organoid cultures were established were excluded for this analysis because the tumor cell percentage of the biopsy was below 30%.

A mean of 19 (±4) somatic mutations were observed per patient (Fig. 2). Ninety percent of somatic mutations were shared between tumor biopsy and organoid cultures. Somatic mutations in driver genes or other genes of interest were invariably detected in both tumor biopsy and biopsy-derived cultures from all patients (Fig. 2). An average correlation of 0.89 (range 0.77–0.96) was detected for DNA copy number profiles between matching tumor biopsy and organoid samples (Fig. 2). The average percentage of data points detected identically as gain, loss, or neutral was 81% (range: 52–99%). Somatic mutations identified are presented in Table S1, sequencing statistics in Table S2, and tumor cell percentage and necrosis percentage of original biopsies in Table S3. Copy number profiles for matched tumor biopsies and biopsy-derived cultures per patient are shown in Fig. S1.

Fig. 2.

Fig. 2.

Copy number and mutational profiles of biopsy-derived organoid cultures and matched tumor biopsies for patients 01–08. (A) Venn diagrams displaying the number of shared and unique (tumor biopsy or biopsy-derived cultures) mutations per patient as identified by SOLiD sequencing. (B) Mutations in known driver genes or genes of interest for colorectal cancer per patient. Mutations were invariably present in both tumor biopsy and biopsy-derived organoid cultures. (C) Heat map comparing copy number aberrations between matched tumor biopsy and biopsy-derived organoid cultures in a log2 scale. Red colors indicate gains, and blue colors indicate losses. The band on the left represents chromosomes 122 and X. For each patient the correlation between the tumor biopsy and biopsy-derived cultures is shown, as well as the percentage of data points detected identically as gain, loss, or neutral (overlap).

Table S1.

SOLiD data somatic mutations

Subject Chromosome Position Base change Amino acid change Mean coverage Gene Variant type Germline coverage Tumor Coverage Organoid coverage Germline allele frequency Tumor allele frequency Organoid allele frequency Shared (S), tumor unique (T), or organoid unique (O)
1 1 209,797,208 T/A K705I 332 LAMB3 SNV 298 459 239 2 85 99 S
2 179,407,388 C/A NA 493,67 TTN SNV 489 634 358 0 20 19 S
2 179,486,728 C/A W6034L 344,33 TTN SNV 253 520 260 0 39 43 S
3 25,665,813 G/A P824S 775,67 TOP2B SNV 694 1127 506 1 22 24 S
3 119,582,332 G/A R357W 401 GSK3B SNV 316 640 247 1 28 39 S
3 195,605,915 C/G NA 45,33 TNK2 SNV 53 71 12 0 20 17 S
4 65,298,248 A/G NA 11,33 RP11-204H9.2 SNV 15 17 2 0 76 100 S
5 112,175,021 C/T Q1244* 96,67 APC SNV 96 133 61 1 21 34 S
6 4,059,028 G/A V514I 57 PRPF4B SNV 82 34 55 1 21 7 S
8 8,999,186 C/T NA 83,33 PPP1R3B SNV 113 119 18 0 68 100 S
11 106,810,698 C/G E232Q 693,33 GUCY1A2 SNV 508 1145 427 0 27 24 S
11 117,306,398 C/A R1673L 301 DSCAML1 SNV 349 390 164 0 31 29 S
12 57,849,972 C/T P132S 180 INHBE SNV 122 323 95 0 21 32 S
13 32,972,674 G/A E3342K 846,67 BRCA2 SNV 490 1434 616 0 17 28 S
15 99,442,705 G/A NA 351 IGF1R SNV 241 603 209 0 26 39 S
16 3,777,892 C/T G2348R 49,67 CREBBP SNV 45 76 28 0 53 68 S
20 2,845,936 C/T A402V 187,33 VPS16 SNV 105 347 110 0 8 19 S
X 41,027,389 C/T R852* 164,67 USP9X SNV 251 118 125 0 79 93 S
17 75,775,39 G/A R248W 205,33 TP53 SNV 235 280 101 0 69 79 S
2 1 10,363,653 G/C E804Q 163,67 KIF1B SNV 128 184 179 0 8 17 S
1 43,814,930 C/G NA 103 MPL SNV 86 127 96 0 17 21 S
2 99,172,162 G/A NA 737,33 INPP4A SNV 606 996 610 0 24 30 S
2 179,486,342 A/G I6005T 899,67 TTN SNV 1422 224 1053 0 16 28 S
2 179,507,003 G/A R4567C 307 TTN SNV 714 55 152 0 44 45 S
3 1,414,000 G/A V504I 375,67 CNTN6 SNV 530 158 439 0 1 16 S
3 112,991,517 G/A A310T 20,67 BOC SNV 7 34 21 0 41 14 S
4 23,825,968 T/G K271T 465 PPARGC1A SNV 793 153 449 0 16 26 S
4 146,467,864 C/T A262V 152 SMAD1 SNV 287 24 145 0 38 42 S
5 112,155,013 C/- NA 150,67 APC Indel 179 155 118 0 60 90 S
5 170,818,803 G/A A118T 240,67 NPM1 SNV 328 179 215 0 0 18 O
6 57,075,028 T/A I51F 474 RAB23 SNV 621 176 625 0 51 43 S
11 92,533,992 T/A Y2605N 783 FAT3 SNV 877 596 876 0 40 41 S
16 77,353,825 G/A T818M 106,33 ADAMTS18 SNV 129 122 68 0 53 72 S
17 7,576,927 C/A NA 144 TP53 SNV 175 124 133 0 64 99 S
19 39,996,053 A/C K352T 60,33 DLL3 SNV 67 70 44 0 13 16 S
3 46,307,365 G/A R260Q 656 CCR3 SNV 693 686 589 0 23 20 S
12 25,398,281 C/T G13D 207,33 KRAS SNV 326 109 187 0 85 91 S
3 1 227,300,460 C/T R601Q 693,67 CDC42BPA SNV 655 587 839 0 14 35 S
4 48,073,695 C/G NA 237 TXK SNV 247 282 182 0 0 18 O
5 112,175,480 G/T E1397* 349 APC SNV 408 289 350 0 2 36 S
6 32,184,739 C/A C615F 133,33 NOTCH4 SNV 104 172 124 1 5 47 S
10 64,968,097 G/C S892C 285,33 JMJD1C SNV 328 204 324 0 24 68 S
10 114,912,204 G/A NA 130,67 TCF7L2 SNV 96 187 109 1 7 32 S
11 117,308,068 C/T R1287Q 87 DSCAML1 SNV 61 137 63 2 15 94 S
11 118,342,652 C/G P293A 446,33 MLL SNV 390 462 487 0 9 39 S
12 120,653,800 T/G K4T 27 PXN SNV 19 49 13* 0 18 0 T
12 121,416,813 T/C F81S 18,33 HNF1A SNV 8 22* 25 0 0 16 O
15 44,843,120 T/C V65A 161,33 EIF3J SNV 192 145 147 0 0 25 O
15 88,690,618 G/A R138W 494,33 NTRK3 SNV 500 490 493 0 26 99 S
17 7,577,095 G/T D281E 192,67 TP53 SNV 160 297 121 0 8 95 S
18 45,371,862 T/C NA 276 SMAD2 SNV 297 230 301 0 20 99 S
19 7,976,387 G/T V377F 39,67 MAP2K7 SNV 42 51 26 5 12 77 S
12 25,398,284 C/G G12A 227,33 KRAS SNV 296 265 121 0 4 26 S
4 1 160,388,795 G/T D66Y 63,33 VANGL2 SNV 56 21 113 0 19 46 S
1 226,566,974 T/A −538- 154,67 PARP1 SNV 170 53 241 4 38 13 S
1 227,216,861 C/T R500H 284,67 CDC42BPA SNV 357 53 444 0 32 39 S
1 228,402,727 C/A Q586K 23,33 OBSCN SNV 26 8* 36 0 0 22 O
2 179,643,820 C/T R1330H 160,33 TTN SNV 301 25 155 0 60 43 S
3 10,070,375 G/A D12N 99,67 FANCD2 SNV 235 39 25 0 36 20 S
5 6,743,939 A/T I161F 96,67 PAPD7 SNV 233 25 32 0 40 72 S
5 38,960,005 C/T E643K 211,33 RICTOR SNV 537 33 64 0 27 42 S
5 112,173,917 C/T R876* 312 APC SNV 591 69 276 0 43 98 S
7 19,156,452 T/A S33C 68 TWIST1 SNV 83 32 89 0 0 17 O
8 2,887,015 C/T E2562K 81,67 CSMD1 SNV 140 6 99 0 50 35 S
8 38,275,833 C/T R357Q 84,33 FGFR1 SNV 108 13 132 1 85 57 S
8 103,307,432 G/A P1347L 628 UBR5 SNV 717 79 1088 0 68 68 S
9 113,496,548 C/T R216W 318,67 MUSK SNV 431 98 427 1 28 22 S
11 11,373,733 G/A R312W 205,67 CSNK2A3 SNV 324 46 247 0 13 33 S
17 40,491,345 C/T R152Q 228,33 STAT3 SNV 408 47 230 0 70 98 S
20 2,842,501 G/A R317H 45,67 VPS16 SNV 58 11 68 0 27 21 S
X 1,404,820 C/T NA 99,67 CSF2RA SNV 101 48 150 0 56 40 S
X 153,592,746 G/A NA 32,67 FLNA SNV 22 9* 67 0 0 19 O
12 25,398,285 C/A G12C 140,67 KRAS SNV 240 56 126 1 41 56 S
17 7,578,406 C/T R175H 31 TP53 SNV 33 5 55 0 80 96 S
5 1 242,016,664 C/T R96* 431 EXO1 SNV 532 385 376 0 17 34 S
2 99,180,106 C/A NA 5,33 INPP4A SNV 1 4 11 0 75 55 S
4 103,446,721 G/A NA 74,33 NFKB1 SNV 94 58 71 0 14 21 S
4 104,070,038 T/A Q1244L 243,67 CENPE SNV 388 226 117 0 22 10 S
5 112,175,621 C/T Q1444* 280 APC SNV 436 226 178 0 36 88 S
6 30,856,718 G/A R40Q 200 DDR1 SNV 187 206 207 2 33 64 S
6 162,683,713 C/T D86N 210,33 PARK2 SNV 220 246 165 0 8 19 S
7 141,424,934 C/T R444C 235 WEE2 SNV 186 275 244 0 23 40 S
10 17,142,212 -/A NA 200,67 CUBN Indel 217 204 181 0 7 18 S
15 64,275,855 C/T R64H 58,33 DAPK2 SNV 54 65 56 0 3 20 S
17 27,208,869 C/T E227K 33 FLOT2 SNV 39 26 34 0 50 94 S
18 4,891,888 G/T D351Y 419 SMAD4 SNV 583 353 321 0 52 100 S
12 25,398,281 C/T G13D 182 KRAS SNV 202 164 180 0 49 95 S
14 21,854,022 G/T P2499H 35,67 CHD8 SNV 31 38 38 0 5 18 S
17 7,578,406 C/T R175H 28,67 TP53 SNV 43 14 29 0 50 97 S
6 1 45,809,000 G/A D307N 125,67 TOE1 SNV 290 8 79 1 25 47 S
2 170,009,480 G/A NA 96 LRP2 SNV 93 32 163 0 38 4 S
3 14,200,245 T/G S343R 127,33 XPC SNV 185 98 99 0 33 17 S
4 114,186,052 G/A NA 28,67 ANK2 SNV 55 7* 24 0 0 29 O
5 112,174,864 AA/- NA 256,33 APC Indel 505 100 164 0 36 43 S
6 32,944,234 T/A L273H 316 BRD2 SNV 473 124 351 0 81 76 S
7 76,111,907 G/- NA 23,33 DTX2 Indel 31 15 24 0 27 12 S
8 41,166,327 C/T A118T 11,67 SFRP1 SNV 10 17 8 0 35 12 S
8 103,307,229 T/G K1390T 542 UBR5 SNV 661 135 830 0 17 0 T
9 71,504,018 T/G F147C 237,33 PIP5K1B SNV 363 51 298 0 0 23 O
9 77,359,089 T/G K1690Q 274,33 TRPM6 SNV 405 96 322 0 0 21 O
18 9,522,319 G/A E289K 246,67 RALBP1 SNV 422 31 287 1 23 3 S
19 1,455,191 C/A L153I 32,67 APC2 SNV 77 10 11 0 50 82 S
19 50,374,886 T/G K182T 104,67 AKT1S1 SNV 91 87 136 0 0 16 O
19 56,042,661 C/T G102D 12,33 SBK2 SNV 6 19 12 0 21 25 S
20 8,639,221 G/C Q244H 355,33 PLCB1 SNV 440 185 441 1 16 7 S
X 591,711 G/A G27R 54,33 SHOX SNV 45 64 54 0 56 20 S
17 7,577,114 C/T C275Y 87,67 TP53 SNV 151 11 101 0 82 68 S
7 1 41,404,534 A/- NA 13,33 RP11-348A7.1 Indel 8 2* 30 0 0 17 O
1 44,154,629 G/A E634K 125,67 KDM4A SNV 134 91 152 1 18 2 S
1 198,225,575 G/C E106Q 232 NEK7 SNV 331 156 209 1 19 42 S
2 148,657,319 C/A S127* 442,33 ACVR2A SNV 760 303 264 0 50 94 S
3 178,916,725 C/T R38C 603,67 PIK3CA SNV 750 459 602 0 54 77 S
3 184,293,701 G/A A314T 60,33 EPHB3 SNV 59 49 73 2 16 1 S
5 112,175,423 C/T Q1378* 330 APC SNV 403 228 359 0 68 91 S
7 98,565,256 T/A Y2476N 169,67 TRRAP SNV 260 64 185 0 75 88 S
8 3,432,515 T/G E433D 148 CSMD1 SNV 256 67 121 0 25 86 S
10 412,306 C/A R726I 548,33 DIP2C SNV 616 414 615 0 21 55 S
10 8,115,818 C/G N388K 353,67 GATA3 SNV 446 314 301 0 6 54 S
10 64,967,678 G/C P1032A 454,67 JMJD1C SNV 659 275 430 0 21 47 S
11 116,719,906 G/A S983F 45 SIK3 SNV 41 37 57 0 22 2 S
11 117,392,003 G/A T412M 107,33 DSCAML1 SNV 127 117 78 0 11 26 S
13 110,804,709 C/T A1634T 141 COL4A1 SNV 122 123 178 1 8 42 S
16 68,842,430 C/A P87Q 207,67 CDH1 SNV 275 126 222 0 6 17 S
4 153,247,288 C/A R505L 583,33 FBXW7 SNV 783 382 585 0 25 46 S
12 25,398,284 C/A G12V 176,33 KRAS SNV 294 111 124 1 13 28 S
17 7,578,406 C/T R175H 25 TP53 SNV 39 27 9 0 26 67 S
8 1 9,793,472 T/C E795G 153,67 CLSTN1 SNV 186 161 114 0 14 46 S
1 12,164,486 G/A E107K 430,67 TNFRSF8 SNV 453 474 365 1 9 38 S
1 165,876,941 C/T Q223* 128 UCK2 SNV 157 113 114 0 13 57 S
2 33,482,484 A/T K441N 344,67 LTBP1 SNV 389 341 304 0 18 34 S
2 121,708,928 G/A V122M 48,33 GLI2 SNV 57 33 55 0 6 29 S
2 171,884,902 C/T E347K 267,67 TLK1 SNV 345 323 135 1 6 22 S
2 179,547,627 A/G V10647A 110,67 TTN SNV 168 77 87 0 14 34 S
2 179,622,344 C/A G3489W 305 TTN SNV 344 299 272 4 12 48 S
2 215,593,455 G/A S131L 597 BARD1 SNV 639 555 597 1 12 38 S
3 1,444,098 A/C I900L 305,67 CNTN6 SNV 429 231 257 0 38 94 S
3 25,659,978 C/A L1089F 140,67 TOP2B SNV 191 136 95 1 3 16 S
3 30,732,969 C/T R528C 49 TGFBR2 SNV 77 34 36 0 29 89 S
3 171,455,712 C/T V44M 134,33 PLD1 SNV 208 77 118 0 10 26 S
5 112,175,675 AG/- NA 288,33 APC Indel 382 257 226 0 11 54 S
7 6,038,819 C/T G209R 590,67 PMS2 SNV 611 612 549 1 12 38 S
8 48,715,930 G/A R3286W 173 PRKDC SNV 224 173 122 1 6 39 S
8 121,282,371 C/A S1057R 190,67 COL14A1 SNV 303 120 149 1 8 31 S
9 94,486,903 C/T D485N 156,67 ROR2 SNV 196 139 135 1 24 47 S
10 81,006,67 C/T S214L 38,67 GATA3 SNV 53 30 33 2 7 39 S
10 89,692,905 G/T R130L 234 PTEN SNV 281 212 209 0 14 46 S
11 108,128,322 A/C N789H 147 ATM SNV 149 125 167 1 28 54 S
12 52,374,912 A/G E195G 352 ACVR1B SNV 352 298 406 1 35 69 S
12 53,686,729 G/A V2046M 52 ESPL1 SNV 46 47 63 0 15 38 S
19 15,276,227 C/T G110E 125,67 NOTCH3 SNV 164 81 132 1 26 42 S
X 44,920,568 G/A NA 55 KDM6A SNV 63 47 55 3 19 0 T
X 71,813,093 C/A G1052V 264,67 PHKA1 SNV 324 265 205 1 15 72 S
7 140,453,136 A/T V28E 422 BRAF SNV 474 395 397 0 6 35 S
10 89,692,904 C/T R130* 233 PTEN SNV 278 212 209 0 16 45 S

Overview of all somatic mutations identified by SOLiD sequencing. Unique mutations are highlighted in boldface. The allele frequency is the percentage of reads in which the mutant allele has been detected. SNV, single-nucleotide variant.

*

Low coverage for samples with a unique mutation (might have resulted in the inability to identify a mutation).

Table S2.

Sequencing statistics depicted separately for all samples from each patient

Sample Total reads % mapped % uniquely mapped % in target bases Average coverage Median coverage % bases covered % bases covered >20×
Subject01 - blood 55,725,994 87.65 82.13 55.57 231.69 174.00 94.55 80.71
Subject01 - organoid 40,839,236 87.72 81.78 58.99 180.51 117.00 93.65 76.53
Subject01 - tumor 89,556,002 88.03 82.11 51.25 344.62 230.00 95.82 85.74
Subject02 - blood 78,031,553 88.48 82.46 55.59 327.50 248.00 95.41 84.21
Subject02 - organoid 57,602,210 87.38 81.41 55.31 237.68 155.00 94.80 81.20
Subject02 - tumor 58,474,524 85.93 80.16 54.31 232.86 157.00 96.19 86.96
Subject03 - blood 60,117,496 87.51 82.05 55.64 249.91 190.00 94.68 81.40
Subject03 - organoid 55,752,800 88.87 83.60 55.66 235.65 160.00 94.75 80.97
Subject03 - tumor 72,388,430 86.26 80.84 55.52 296.03 248.00 96.07 87.57
Subject04 - blood 59,027,598 86.57 80.78 55.33 241.00 189.00 95.30 83.18
Subject04 - organoid 42,977,631 89.45 84.03 56.05 184.11 106.00 94.36 78.99
Subject04 - tumor 20,034,396 79.87 73.20 41.30 56.46 31.00 89.01 59.71
Subject05 - blood 58,658,789 87.12 81.22 53.58 233.41 181.00 94.89 81.71
Subject05 - organoid 45,661,474 88.40 82.37 54.46 187.51 144.00 94.87 80.89
Subject05 - tumor 51,527,869 88.07 82.24 56.14 217.18 157.00 94.57 81.14
Subject06 - blood 62,923,615 79.67 74.27 54.87 234.91 195.00 94.68 83.73
Subject06 - organoid 24,908,884 68.34 62.27 45.03 65.48 42.00 94.45 70.95
Subject06 - tumor 61,725,207 63.98 59.68 56.61 190.95 132.00 93.63 79.90
Subject07 - blood 66,097,552 82.22 76.95 58.13 269.70 226.00 95.15 85.49
Subject07 - organoid 65,525,831 79.34 74.36 59.96 266.39 201.00 94.99 84.86
Subject07 - tumor 41,859,372 76.29 71.42 60.34 164.69 132.00 94.54 82.40
Subject08 - blood 57,698,769 78.87 73.78 59.47 231.04 188.00 95.19 84.77
Subject08 - organoid 55,896,268 75.59 70.73 60.17 217.12 162.00 94.67 82.34
Subject08 - tumor 49,018,897 76.00 71.18 60.87 193.64 156.00 94.68 82.79

Table S3.

Percentage of tumor cells and necrosis of original tumor biopsies per patient

Sample Tumor cells, % Necrosis, %
01 80 10
02 90 10
03 80 40
04 90 10
05 70 10
06 90 50
07 80 50
08 90 Not available

Fig. S1.

Fig. S1.

Copy number plots for tumor biopsy and biopsy-derived organoid cultures of patients 01–08.

Discussion

These data show that organoid cultures from colorectal cancer can be established readily from biopsies of metastases. The culture success rate of 71% for colorectal cancer compares favorably with the take rate of 15–20% for organoid cultures of biopsies from prostate cancer described by Gao et al. (6). Sequencing data and copy number analysis were largely concordant between organoid cultures and tumor biopsies. The most important observation from our study, however, is that none of the 15 mutations that were found exclusively in either the tumor or organoid culture are in driver genes or genes amenable for drug targeting. Discordance, when present, could be attributed to biological factors such as sampling variation from a heterogeneous tumor or ongoing normal tumor evolution or to selection resulting from culture conditions. With current knowledge it is not possible to distinguish between these factors. Aside from biological processes, technical limitations can result in a misrepresentation of the actual concordance by impeding factors such as low coverage of a particular gene or sample. For instance, 6 of 15 unique mutations have a coverage of <25× of the gene in the matched sample (where the mutation was not identified), and for patient 04 and patient 06 overall coverage of the tumor sample and biopsy-derived culture, respectively, was very low (median coverage <50×) (Tables S1 and S2). Furthermore, the presence of stromal cells and tumor necrosis in biopsies (resulting in a lower tumor percentage) may have resulted in less accurate detection of copy number aberrations and somatic mutations in the tumor biopsy as compared with the cultures. A lower tumor percentage can result in a misrepresentation of allele frequency in a negative way, so that the frequency can fall under the limit of detection. The actual allele frequency of a mutation in tumor biopsies thus is sometimes higher than depicted in Table S1. A lower tumor percentage also affects copy number profiles by reducing the signal toward the diploid state, resulting in discrepancies in the intensity of red and blue shading between tumor biopsies and corresponding biopsy-derived cultures in Fig. 2. This effect could have contributed to the discordance in patient 3 (Fig. S1 and Table S3).

To conclude, organoid cultures from colorectal cancer can be established readily from biopsies of metastases with preservation of the mutational and copy number landscape. This result supports the translation of patient-derived tumor organoids to the clinic as an ex vivo test platform, with the potential to meet the dire need for a means to make more rational treatment decisions for the individual patient. Clinical trials to validate the predictive value of organoids have been initiated.

Acknowledgments

We thank Marja van Blokland (Molecular Pathology, University Medical Center Utrecht) for DNA isolation and Nicolle Besselink (Medical Oncology, University Medical Center Utrecht) for library preparation and sequencing. This work was supported by the Center for Personalized Cancer Treatment, the Barcode for Life Foundation, and Cancer GenomiCs.nl through The Netherlands Organization for Scientific Research Gravitation grant.

Footnotes

The authors declare no conflict of interest.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1516689112/-/DCSupplemental.

References

  • 1.Burstein HJ, et al. American Society of Clinical Oncology American Society of Clinical Oncology clinical practice guideline update on the use of chemotherapy sensitivity and resistance assays. J Clin Oncol. 2011;29(24):3328–3330. doi: 10.1200/JCO.2011.36.0354. [DOI] [PubMed] [Google Scholar]
  • 2.Hidalgo M, et al. A pilot clinical study of treatment guided by personalized tumorgrafts in patients with advanced cancer. Mol Cancer Ther. 2011;10(8):1311–1316. doi: 10.1158/1535-7163.MCT-11-0233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Sato T, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141(5):1762–1772. doi: 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
  • 4.Sato T, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459(7244):262–265. doi: 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
  • 5.Barker N, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6(1):25–36. doi: 10.1016/j.stem.2009.11.013. [DOI] [PubMed] [Google Scholar]
  • 6.Gao D, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176–187. doi: 10.1016/j.cell.2014.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Huch M, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 2013;32(20):2708–2721. doi: 10.1038/emboj.2013.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Huch M, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494(7436):247–250. doi: 10.1038/nature11826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Karthaus WR, et al. Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell. 2014;159(1):163–175. doi: 10.1016/j.cell.2014.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.van de Wetering M, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–945. doi: 10.1016/j.cell.2015.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Harakalova M, et al. Multiplexed array-based and in-solution genomic enrichment for flexible and cost-effective targeted next-generation sequencing. Nat Protoc. 2011;6(12):1870–1886. doi: 10.1038/nprot.2011.396. [DOI] [PubMed] [Google Scholar]
  • 12.Hoogstraat M, et al. Genomic and transcriptomic plasticity in treatment-naive ovarian cancer. Genome Res. 2014;24(2):200–211. doi: 10.1101/gr.161026.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Vermaat JS, et al. Primary colorectal cancers and their subsequent hepatic metastases are genetically different: Implications for selection of patients for targeted treatment. Clin Cancer Res. 2012;18(3):688–699. doi: 10.1158/1078-0432.CCR-11-1965. [DOI] [PubMed] [Google Scholar]
  • 14.Kuilman T, et al. CopywriteR: DNA copy number detection from off-target sequence data. Genome Biol. 2015;16:49. doi: 10.1186/s13059-015-0617-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.van de Wiel MA, Picard F, van Wieringen WN, Ylstra B. Preprocessing and downstream analysis of microarray DNA copy number profiles. Brief Bioinform. 2011;12(1):10–21. doi: 10.1093/bib/bbq004. [DOI] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES