Therapy for Metastatic Gastrointestinal Stromal Tumor


Author, Year

Patients

TKI

R0/R1 rate

Recurrence/PFS

Survival

Predictors

Raut (2006) [68]

23 SD/PR

32 localized progression

14 generalized progression

All

SD/PR: 78 %

LP: 25 %

GP: 7 %

SD/PR: 80 %

LP: 33 %

GP: 0 %

12-month OS:

SD/PR: 95 %

LP: 86 %

GP: 0 %

Responsiveness to TKI

Rutkowski (2006) [69]

24 SD/PR

8 progressive disease

All

SD/PR: 24/24

PD: 5/8

Median time to recurrence: 17 month (SD/PR) vs 12 month (PD)

N/A

In first 5 responders, imatinib was not continued postoperative and 4 recurred. In next 19, only 1 recurrence in median follow-up of 12 months

Bonvalot (2006) [70]

12 planned metastasectomy

5 local advanced

5 emergencies

12/12

10/12

Median: 23.4 month (included 5 locally advanced)

2-year OS: 62 % (all patients)
 
Gronchi (2007) [71]

35 total

27 responding to TKI, 8 not

All

Responding: 24/27

Not responding: 4/8

12-month PFS:

96 % vs 0 %

24-month PFS: 69 %

12-month DSS:

100 % vs 60 %

TKI responsiveness

DeMatteo (2007) [72]

20 SD/PR

13 focal progression

7 multifocal progression

All

SD/PR: 17/20

FP: 6/13

MFP: 2/7

2-year PFS for SD/PR: 61 %

Median TTP: 12 months for FP, 3 months for MFP

2-year OS:

SD/PR: 100 %

FR: 36 %

1-year OS for MFR: 36 %

TKI responsiveness

Andtbacka (2007) [73]

35 recurrent/met

11 locally advanced

All

Recurrent: 11/35

Locally advanced: 11/11

Median time to recurrence: 15.1 month

All R0 alive at 30.7 months. Median OS for incomplete: 12.0 months

Initial responsiveness to imatinib predicted complete resection (91 % vs 4 %)

Al-Batran (2007) [74]

9 FP

16 GP

All

FP: 9/9; GP got dose escalation

11.3 month (FP) vs 2.5 month (GP)

Median OS: not attained (FP) vs 22.8 month (GP)
 
Yeh (2010) [75]

14 PR/SD

21 LP

3 GP

All

PR/SD: 42.9 %

LP: 4.8 %

GP: 0 %

2-year PFS

PR/SD: 59.4 %

LP: 35.9 %

GP: 0 %

2-year OS

PR/SD: 69.6 %

LP: 48.4 %

GP: 0 %

Trend toward more secondary mutations in KIT exon 17 in pts with LP vs SD

Mussi (2010) [76]

49 PR/SD

31 FP

All

PR/SD: 88 % FP: 45 %

2-year PFS:

PR/SD: 64.4 %

FP: 9.7 %

5-year DSS: 82.9 % vs 67.6 %
 
Zaydfudim (2012) [77]

54 surgery

33 no surgery

32/54 preop

35/54

1-year PFS:

91 % PR

58 % SD

11 % DP

1-year OS: 98 % surgery vs 80 %

OS and PFS in surgical group associated with TKI response and R0 resection. Metastasectomy better than medical therapy alone (5-year OS: 65 % vs 11 %)

Bauer (2014) [78]

239

All

177 R0/R1

62 R2

Median PFS: 6.3 years vs 3.4 years

Median OS: 8.7 years vs 5.3 years

Female gender, short interval of imatinib prior to surgery, R0/R1 resection, nonprogressive disease preop, and liver mets

Park (2014) [79]

42 surgery

92 only TKI

All

Surgery: 62 %

Median PFS: 87.7 month vs 42.8 month

Median OS: not reached vs 88.8 month

Surgery group were younger, fewer peritoneal mets

Rubio-Casadevall (2015) [80]

27 surgery while PR/SD

20 surgery for PD

124 no surgery

All

PR/SD: 20/27

PD: 9/20

Median PFS: 73.4 month group 1 vs 44.6 month groups 2/3

Med OS: 87.6 months vs 59.9 month

Improved OS associated with: ECOG performance status, disease limited to one metastatic organ, metastasectomy


Abbreviations: TKI tyrosine kinase inhibitor, PFS progression-free survival, OS overall survival, PR partial response, SD stable disease, LP limited progression, GP generalized progression, PD progressive disease, DSS disease-specific survival, FP focal progression, MFP multifocal progression, R0 macroscopically complete resection with negative microscopic margins, R1 macroscopically complete with positive microscopic margins, R2 macroscopically incomplete, ECOG Eastern Cooperative Oncology Group



Raut et al. [68] published the first large study reporting survival rates of patients with metastatic or locally advanced GIST who had surgery after TKI therapy. They divided patients into three groups based on response to TKI. Twenty-three patients had PR or SD that was deemed completely resectable. Thirty-two patients had localized progression but, importantly, the tumors that did progress were resectable. Fourteen patients had metastatic disease with generalized or multifocal progression. After surgery, there was no evidence of disease in 78 %, 25 %, and 7 % of patients, respectively. The 12-month PFS was 80 %, 33 %, and 0 %, respectively. The 12-month OS was 95 %, 86 %, and 0 %, respectively. Outcomes of surgery and survival rates correlated with responsiveness to TKI therapy. Furthermore, the investigators concluded that patients with advanced GIST that exhibited SD or limited progression with resectable disease benefited from surgical resection.

Gronchi et al. [71] similarly showed in 38 patients with advanced GIST the importance of preoperative response to TKI therapy. In their cohort, 27 patients underwent metastasectomy while responding to imatinib. PFS was 96 % at 12 months and 69 % at 24 months among responders after surgery. The disease-specific survival (DSS) was 100 % at 12 months for responders. In comparison, the nonresponders all progressed by 12 months with DSS of 60 %.

DeMatteo et al. [72] analyzed 40 patients with metastatic GIST who underwent surgery at their center following preoperative imatinib. Patients were categorized as having responsive disease, focal resistance (one tumor growing), or multifocal resistance (multiple tumors growing). The 20 patients with responsive disease had 2-year PFS of 61 % and OS of 100 %. The 13 patients with focal resistance had a median time to progression of 12 months and 2-year OS of 36 %. The seven patients with multifocal resistance had a median time to progression of 3 months and a 1-year OS of 36 %.

Rutkowski et al. [69] reported the results of cytoreductive surgery performed on 32 patients with advanced GIST. Of these patients, 24 had complete or partial response on imatinib while 8 patients had progression, and surgery was performed as salvage therapy. All responders had R0/R1 resections. The first five patients in their series did not resume postoperative imatinib, and four recurred. The subsequent 19 patients received adjuvant imatinib, and only 1 patient recurred. Of the eight patients who did not respond to neoadjuvant TKI therapy, only two patients had R0/R1 resections and five progressed at a median of 12 months; one patient died perioperatively. When initial responders were separated by postoperative imatinib therapy, significant improvement was seen in PFS compared to initial nonresponders.

Bauer et al. [78] published the largest series to date, reporting multi-institutional EORTC-STBSG data. They examined 239 patients who underwent metastasectomy while on TKI therapy; 177 achieved R0/R1 resections, while 62 had R2 resections. The median OS was 8.7 years versus 5.3 years, respectively. The median PFS following R0/R1 resection was 6.3 years compared to 3.4 years for patients undergoing R2 resection. On multivariate analysis of OS, female gender, short interval of imatinib prior to surgery (25 months vs 8 months), R0/R1 resection, nonprogressive disease preoperatively, and liver metastases were positive prognostic factors. Incomplete resection and tumor debulking did not yield survival benefits.

Indications for resection per NCCN guidelines are as follows [19]:


  1. 1.


    Disease that is stable on or responding to TKI therapy when complete gross resection is possible (stable/responsive disease)

     

  2. 2.


    Isolated clones progressing on TKI therapy after initial response (indicative of secondary drug resistance), while other sites of disease remain stable (limited disease progression)

     

  3. 3.


    Emergencies including hemorrhage, perforation, obstruction, or abscess

     

The timing of metastasectomy is not standardized, although most experts agree with initiating TKI therapy and considering surgery depending on disease response around 6 months after TKI initiation. An et al. retrospectively reviewed primary cytoreduction versus imatinib therapy [81]. They compared 35 patients who underwent surgical cytoreduction of >75 % of tumor bulk prior to starting imatinib to 214 patients who started imatinib without surgery and showed no improvement in prognosis with cytoreduction first. Verweij et al. showed in their randomized trial comparing 400 mg of imatinib to 800 mg that the median time to best response is 3.5 months and that there is minimal incremental tumor shrinkage after 9 months [11]. Fairweather et al. recommend operating between 6 and 12 months or at a point when there is no significant change between staging CTs [67]. NCCN guidelines recommend discussing surgery after 6–12 months of disease stability on TKI therapy [19]. Despite data from the multiple retrospective studies above, cytoreductive surgery following TKI therapy has not been shown to be superior to TKI therapy alone. This can only be answered by a randomized clinical trial; attempts at such a trial have failed due to poor accrual. Therefore, at this point, metastasectomy may be beneficial in selected individuals, but patients should be counseled that there are no data to prove it will improve survival over staying on TKI therapy alone.

One retrospective study by Raut et al. [82] reviewed 50 patients with metastatic GIST who underwent surgery following sunitinib treatment to determine if the responsiveness to sunitinib was associated with patient outcomes. Complete gross resection (R0/R1) was achieved in half of patients. The completeness of resection, PFS, and OS were not significantly correlated with response to sunitinib, likely reflecting selection biases in identifying appropriate patients for surgery on sunitinib.

In general, resection appears to benefit patients who have a PR or SD, and possibly those with isolated sites of progression. If there is isolated progression of a few individual lesions on surveillance, resection is reasonable, although there have been no studies to evaluate this versus changing TKI therapy. Surgery was generally not helpful in patients who had generalized progression on TKI therapy. When all metastatic sites can be resected, TKI therapy may prolong disease-free intervals. Incomplete resection may still potentially prolong progression-free intervals by removing drug-resistant clones; disease-free interval may still be prolonged by TKI therapy as long as the remaining disease remains drug responsive.

Importantly, surgery is not an alternative to TKI therapy. TKI therapy is generally continued until surgery (stopping imatinib 24 h, sunitinib 72 h, or regorafenib 1 week prior to surgery), and all patients undergoing surgery should resume drug therapy postoperatively indefinitely as soon as able to tolerate an oral diet.



5.2 Liver Resection


The liver is the site of metastasis in approximately 65 % of patients with relapsed GIST [83]. A few studies have examined outcomes following resection of liver metastases prior to the imatinib era. DeMatteo et al. [84] looked at 34 patients with either GIST or gastrointestinal leiomyosarcoma who underwent complete resection of hepatic metastases and found a median survival of 38 months, with 30 % of patients alive at 5 years. Nunobe et al. [85] found similar results in their series of 18 patients who underwent hepatectomy for metastatic GIST with a median survival of 36 months and 5-year survival of 34 %. Finally, Shima et al. [86] showed in a series of ten patients a median survival of 39 months following hepatectomy. Of note, de la Fuente et al. [87] compared 43 patients with isolated liver metastases (34 underwent surgery) to 16 patients with liver and peritoneal disease (13 underwent surgery). Patients with isolated liver metastases undergoing surgery trended toward longer OS (40.5 months vs 28.7, p = 0.620) and trended toward having a lower recurrence rate after surgery (16/34 vs 8/13 p = .08), suggesting having liver-only disease may be a predictor of slightly better prognosis. This was further confirmed in long-term follow-up of EORTC 62005 study [72]. In patients with only liver metastases, median OS was not reached compared to 7 years in patients with only peritoneal metastases and 3.7 in patients with both. Interestingly, in the group of patients who were operated on while in remission and achieved a complete macroscopic resection, median OS was not reached again in liver-only group, 8.7 years in peritoneal-only group, and increased to 8.1 years in patients with both.

After the advent of imatinib, it has been shown that the combination of imatinib and hepatic resection can offer long-term disease control in patients with isolated hepatic metastases in multiple studies (Table 2).


Table 2
Selected series reporting outcomes after multimodality therapy for GIST liver metastases



































































Author

Patients

Preop TKI

Postop TKI

R0 rate

DFS

OS

Factors

Pawlik (2006) [88]

36

15

11

N/A

1 year: 52 %

3 year 21 %

5 year 16 %

1 year 91 %

3 year 65 %

5 year 27 %

Adjuvant TKI led to longest median OS

Xia (2010) [89]

19

19

19

N/A

N/A

3-year OS: 89.5 % in surgical vs 60 % in nonsurgery groups

Significant OS benefit from surgery. Surgery also improved OS in patients who responded poorly to preoperative TKI

Turley (2012) [90]

39

19

27

92 %

1 year 63 %

3 year 34 %

5 year 26 %

1 year 97 %

3 year 67 %

Surgery and postoperative TKI improve survival

Cananzi (2014) [91]

11

11

9

64 %

1 year 87 %

2 year 62 %

1 year 81 %

2 year 71 %

R0 resection and clinical TKI response correlated with OS

Brudvik (2015) [92]

49

39 perioperative

39 perioperative

47/49

5 year 35.7 %

47.1 % with TKI vs 9.5 % without

5 year 55.3 %

10 year 52.5 %

Imatinib improved survival


Abbreviations: TKI tyrosine kinase inhibitor, DFS disease-free survival, OS overall survival, R0 macroscopically complete resection with negative microscopic margins

Turley et al. [90] studied 39 patients that underwent hepatic resection for metastatic GIST between 1995 and 2010. Thirty-one patients received TKI therapy. Three-year OS in patients receiving TKI was 71.9 % and 0 % in the seven patients who did not receive TKI therapy. They also noted that patients who were exposed to TKI therapy for prolonged periods (median 18 months) preoperatively trended toward worsened OS, suggesting against delaying surgery indefinitely. Their median OS exceeded previous reports for treatment of metastatic GIST with hepatic resection alone of 36–47 months [8486, 88].

Xia et al. [89]reported 39 patients with metastatic GIST to the liver treated with either 6 months of neoadjuvant imatinib followed by surgery and 2–4 weeks of adjuvant therapy or imatinib alone. The 3-year OS was 89.5 % in the surgery group and 60 % in the imatinib-alone group, which was significant. Also significant was that among patients who responded poorly to 6 months of preoperative imatinib, surgery significantly improved OS compared to those that did not undergo surgery (p = 0.04)

Zhu et al. [93] studied 42 patients with recurrent GIST to either the liver or abdomen treated with long-term imatinib alone without surgery. They found a combined median OS of 48 months. Notably, the median time to progression was longer in the liver group (48 months versus 39 months in patients with only abdominal metastases and 33 months in patients with both), but this was not significant. Median OS was not reached in the liver-alone group with three of the ten patients dying during over 3 years of follow-up. This showed that imatinib alone is a reasonable option in patients with hepatic metastases as well as other metastatic disease.

The NCCN [19] and ESMO [94] both recommend indefinite adjuvant imatinib for patients with resected liver metastases, even if resection was complete.



6 Surveillance


There presently is no standardized follow-up regimen for patients with GIST. The NCCN guidelines [19] recommend an abdominal/pelvic CT be obtained every 3–6 months after surgical resection of GIST and within 3 months after initiating TKI therapy for patients with advanced GIST. ESMO guidelines [94] also recommend follow-up CT or MRI every 3–6 months in patients receiving adjuvant therapy for the first 3 years. Upon cessation of adjuvant therapy, ESMO further recommends follow-up imaging every 3 months for the first 2 years followed by less frequent imaging if stable.

CT scans are the typical initial imaging modality of choice. GISTs typically appear as a solid contoured mass that enhances brightly with IV contrast on CT scans. Larger tumors can be less homogenous due to hemorrhage and necrosis within the tumor. MRI can be useful in patients that cannot receive IV contrast for CT scans. MRIs are also better at evaluating GISTs in the liver and rectum [95], which is especially important ahead of a planned surgery.

PET scans are highly sensitive for detecting GISTs, but lack specificity. While not a stand-alone imaging modality for surveillance, PET can be used for detecting an unknown primary site or resolving ambiguities from CT [16]. Another potential scenario where PET can be considered is when response to therapy must be determined quickly. Specifically, TKI responsiveness can be seen as early as 1 day after treatment is started on PET scan compared to 1–2 months on standard CT scans [9698].

GISTs that respond to TKI therapy become more homogenous and hypodense, and sometimes will later shrink in size as well. This is important when considering the two major criteria systems for GIST follow-up: RECIST and Choi. RECIST (Response Evaluation Criteria In Solid Tumors) was developed first. which determines treatment response based upon tumor measurements [99].However, it was known that this was not necessarily well-suited for GIST surveillance, as early response to TKI therapy often does not correlate with decrease in tumor size, and further signs of tumor progression on TKI therapy are often seen as new areas of hyperdensity before an increase in size. This led Choi et al. [100] to develop a different response evaluation system that used both tumor density and size (Table 3).


Table 3
Choi criteria for tumor responsiveness in GIST






















Response

Definition

Complete response

1. Disappearance of all lesions

2. No new lesions

Partial response

1. A decrease in size of 10 % or more; or a decrease in tumor density (HU) of 15 % or more on CT

2. No new lesions

3. No obvious progression of nonmeasurable disease

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 30, 2017 | Posted by in GASTOINESTINAL SURGERY | Comments Off on Therapy for Metastatic Gastrointestinal Stromal Tumor

Full access? Get Clinical Tree

Get Clinical Tree app for offline access