1,121
Views
11
CrossRef citations to date
0
Altmetric
Commentary on selected article in this issue

A “PET” topic in primary mediastinal large B-cell lymphoma: positive or negative, and how to handle it in the end

, &

Primary mediastinal large B-cell lymphoma (PMLBCL) is regarded as a separate entity among the aggressive large B-cell lymphomas, characterized by unique clinical, pathologic and molecular features [Citation1]. Immunochemotherapy with rituximab–cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP) is still viewed as the current standard of care, but several promising more aggressive regimens have also been used in this disorder [Citation2–6]. However, until now these alternative combination chemotherapy regimens have not yet been directly compared systematically [Citation2–6]. Radiotherapy is frequently used as an additional treatment modality for this lymphoma, but its precise role still needs to be further defined. Since progression-free survival (PFS) for PMLBCL after therapy is now 75–90% and long-term overall survival 85–90%, earlier identification of the minority of patients who are destined to fail treatment is important [Citation2–6]. In this respect it is possible that utilizing imaging techniques such as positron emission tomography–computed tomography (PET-CT) during the course of the disease and at the end of therapy may provide important data, which could help in recognizing a subgroup of patients who are most likely to fail therapy.

PET-CT is increasingly used in Hodgkin lymphoma (HL) and diffuse large B-cell lymphoma (DLBCL) [Citation7–9]. In the interim setting, PET-CT (iPET) may facilitate early treatment modification, while end-of-treatment PET-CT (ePET) may well guide decisions regarding the addition of complementary radiotherapy or further salvage therapy. Until now, such decisions have essentially been based on empirical data, since the results of large ongoing randomized trials are still awaited. However, in the specific setting of PMLBCL, both iPET and ePET are widely used in daily practice, based merely on projection of data and conclusions derived from prior clinical experience gained with HL and DLBCL.

In this issue of the journal, Cheah et al., from Peter McCallum Cancer Center in Melbourne, Australia, report their single-institution experience on the use of PET-CT for interim and final evaluation of response in 28 patients with PMLBCL [Citation10]. In this series R-CHOP-14 or R-CHOP-21 were the most frequently used regimens (n = 21; no R in one case), while only four and three patients received R-da-EPOCH (R-dose adjusted etoposide, prednisolone, vincristine, cyclophosphamide, doxorubicin) and Hyper-CVAD ± R (hyperfractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, with or without R), respectively.

Regarding iPET (mostly performed after two or three cycles), 35% of 23 patients with available data were reported as positive according to the standard Deauville criteria (score 4 or 5) [Citation11], while 30% had positive findings when evaluated according to a modified version of the International Harmonization Project (IHP) criteria [Citation12,Citation13]. Irrespective of the method used, iPET appeared to have little impact on the eventual patient outcome. Nevertheless, in this study chemotherapy was escalated in some patients in response to receiving a report of a positive iPET (iPET-pos), with a potential further reduction of the positive predictive value (PPV), which was only 12–14% in this series. This observation is generally in keeping with other studies: Moskowitz et al. reported the results of iPET after four cycles of chemotherapy in 51 patients treated with augmented R-CHOP-14 × 4 followed by additional consolidation therapy with three cycles of ICE (ifosfamide, carboplatin, etoposide) chemotherapy, in cases who were iPET-negative (iPET-neg) or iPET-pos but with a negative biopsy [Citation14,Citation15]. The minority of iPET-pos patients, who had a positive biopsy, received a similar consolidation regimen plus R, followed by autologous stem cell transplant (ASCT). As iPET positivity was more loosely defined here as uptake > background, 47% of patients were regarded as iPET-pos. After the administration of a non-cross-resistant subsequent treatment, iPET had no impact on the outcome at all, and using a change in maximum standardized uptake value (ΔSUVmax)-based criterion was also shown to be insufficient in this respect [Citation15]. In another recently published study, Avigdor et al. analyzed iPET results in 30 patients, mostly treated with R-VACOP-B (R–etoposide, doxorubicin, cyclophosphamide, vincristine, prednisone, bleomycin) (R-CHOP-21 was also used but to a lesser extent) without radiotherapy [Citation16]. In this series, 47% of the patients were regarded as iPET-pos using the IHP criteria, and 5-year PFS for iPET-neg vs. -pos patients was 94% vs. 57%, respectively. Although this difference was statistically significant, its magnitude was much less impressive than that observed in HL [Citation8]. However, in this respect, it should be stressed that very heterogeneous definitions for iPET positivity were used in the different studies [Citation10,Citation14–16]. Based on the above data, current evidence for the routine use of iPET in PMLBCL outside clinical trials still appears to be too inconclusive to recommend it definitively as a routine procedure.

The most interesting and instructive part of the study reported here by Cheah and colleagues relates to the implications of ePET results on the outcome and further management of patients with PMLBCL [Citation10]. Using the same criteria for positivity as for iPET, the authors set a much more stringent than the usual definition for end-of-treatment PET positivity [Citation10–13], and as a result, ePET was considered positive only if the Deauville score was 4 or 5 (but not 3) or if a modified IHP criterion was met. Under this strict definition, only seven of 28 (25%) patients remained as ePET-pos and, while the negative predictive value (NPV) for ePET was 95%, the positive predictive value (PPV) was only 43%. These seven patients and the two patients with Deauville score 3 were not irradiated but received salvage therapy with R-ICE or Hyper-CVAD and ASCT (in 8/9), although 5/9 had achieved a conventional radiographic response and no biopsy was performed prior to the initiation of salvage therapy. After ASCT, six of these nine patients remained in remission, and this is the reason for the low PPV of ePET. It is also of interest that among four mediastinal biopsies performed after ASCT because of persistent PET positivity, three were negative, revealing foamy macrophages and/or necrotic changes.

Recent moderately sized or large studies have attempted to address the issue of ePET in PMLBCL [Citation5,Citation17–21]. Using less strict criteria (either the IHP or Deauville score ≥ 3), 41–69% of patients had a positive ePET. When radiotherapy was applied uniformly to ePET-pos patients, a positive ePET was associated with inferior PFS, but the magnitude of the difference between ePET-neg and -pos patients was < 25–30% [Citation18,Citation20], and statistical significance was borderline or even absent in some studies [Citation17,Citation19,Citation21]. Generally, ePET-pos patients enjoyed long-term PFS of > 65% if given radiotherapy, illustrating that this was adequate treatment for most patients, and leaving salvage chemotherapy and ASCT as a questionable option. However, in patients with more intense fluorodeoxyglucose (FDG) uptake, the efficacy of radiotherapy appeared to be more limited, as most relapses or disease progressions occurred in responding patients with Deauville scores of 5 (without primary progression) or 4 or in those with SUVmax ≥ 5 (roughly corresponding to Deauville score 5 or the highest level of uptakes for Deauville 4) [Citation17,Citation18,Citation20]. Furthermore, according to personal experience (unpublished data) and some National Cancer Institute (NCI) data as well [Citation5], several ePET-pos patients still remain PET-pos after radiotherapy without experiencing relapse in the long-term. This observation may also support the possibility of false positive ePET interpretations in PMLBCL, especially if FDG uptake is relatively low. Finally, it is noteworthy that the above observations appear valid irrespective of the chemotherapy regimen used, i.e. R-CHOP [Citation17–19], R-MACOP-B (R–methotrexate, doxorubicin, cyclophosphamide, vincristine, prednisone, bleomycin)/R-VACOP-B [Citation17,Citation20,Citation21] or R-da-EPOCH [Citation5]. Thus in our view, the conclusion drawn by Cheah et al. [Citation10] appears to be extremely valid, clinically relevant and worth stressing again: “Positive ePET scan by visual analysis or 5PS [5-point scale] predicts inferior PFS, but an inflammatory response resulting in metabolic abnormality is frequently observed and is difficult to differentiate from residual disease. Metabolically active residual masses after the completion of treatment should be biopsied to confirm viable lymphoma if salvage therapy is planned.” [Citation10].

In conclusion, there is currently no convincing evidence for the use of interim PET-CT in PMLBCL. On the other hand, at the end of immunochemotherapy, PET-CT is a valuable tool for the evaluation of response. The ongoing International Extranodal Lymphoma Study Group (IELSG)-37 study will, in the near future, no doubt reveal whether radiotherapy can be omitted in cases with a negative ePET [Citation22]. However, in the mean time, it is important to recognize that PET-CT positivity at the end of therapy is not universally an indication for salvage therapy and ASCT. In this regard, it may be prudent to suggest that only patients with radiographic progression, as documented by conventional imaging, should proceed to ASCT. In contrast, the majority of ePET-pos patients, i.e. those who have achieved at least a partial response, should receive radiotherapy or follow a biopsy-driven approach to achieve satisfactory final outcomes.

Supplemental material

ilal_a_929674_sm2385.zip

Download Zip (1.4 MB)

Potential conflict of interest

Disclosure forms provided by the authors are available with the full text of this article at www.informahealthcare.com/lal.

References

  • Gaulard P, Harris NL, Pileri SA, et al. Primary mediastinal (thymic) large B-cell lymphoma. In: Swerdlow SH, Campo E, Harris NL, et al., editors. World Health Organization classification of tumors of haematopoietic and lymphoid tissues. 4th ed. Lyon: IARC Press; 2008. pp 250– 251.
  • Rieger M, Osterborg A, Pettengell R, et al. Primary mediastinal B-cell lymphoma treated with CHOP-like chemotherapy with or without rituximab: results of the Mabthera International Trial Group study. Ann Oncol 2011;22:664–670.
  • Vassilakopoulos TP, Pangalis GA, Katsigiannis A, et al. Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone with or without radiotherapy in primary mediastinal large B-cell lymphoma: the emerging standard of care. Oncologist 2012;17:239–249.
  • Zinzani PL, Stefoni V, Finolezzi E, et al. Rituximab combined with MACOP-B or VACOP-B and radiation therapy in primary mediastinal large B-cell lymphoma: a retrospective study. Clin Lymphoma Myeloma 2009;9:381–385.
  • Dunleavy K, Pittaluga S, Maeda LS, et al. Dose-adjusted EPOCH-rituximab therapy in primary mediastinal B-cell lymphoma. N Engl J Med 2013;368:1408–1416.
  • Pohlen M, Gerth HU, Liersch R, et al. Efficacy and toxicity of a rituximab and methotrexate based regimen (GMALL B-ALL/NHL 2002 protocol) in Burkitt's and primary mediastinal large B-cell lymphoma. Am J Hematol 2011;86:61–64.
  • Vassilakopoulos TP, Prassopoulos VK. Clinical implications of the role of 18FDG-PET/CT in malignant lymphomas. In: Gouliamos AD, Andreou J, Kosmidis P, editors. Imaging in clinical oncology. Milan: Springer-Verlag Italia; 2014. pp 249–265.
  • Gallamini A, Kostakoglou L. Interim FDG-PET in Hodgkin lymphoma: a compass for a safe navigation in clinical trials? Blood 2012;120:4913–4920.
  • Dührsen U, Hüttmann A, Jöckel KH, et al. Positron emission tomography guided therapy of aggressive non-Hodgkin lymphomas– the PETAL trial. Leuk Lymphoma 2009;50:1757–1760.
  • Cheah CY, Hofman MS, Seymour JF, et al. The utility and limitations of 18F-fluorodeoxyglucose positron emission tomography with computed tomography in patients with primary mediastinal B-cell lymphoma: single institution experience and literature review. Leuk Lymphoma 2015;56:49–56.
  • Meignan M, Gallamini A, Itti E, et al. Report on the third international workshop on interim positron emission tomography in lymphoma held in Menton, France, 26-27 September 2011 and Menton 2011 consensus. Leuk Lymphoma 2012;53:1876–1881.
  • Juweid ME, Stroobants S, Hoekstra OS, et al. Use of positron emission tomography for response assessment of lymphoma:consensus of the Imaging Subcommittee of International Harmonization Project in Lymphoma. J Clin Oncol 2007;25:571–578.
  • Cheson BD, Pfistner B, Juweid ME, et al. Revised response criteria for malignant lymphoma. J Clin Oncol 2007;25:579–586.
  • Moskowitz CH, Schoder H, Teruya-Feldstein J, et al. Risk-adapted dose-dense immnochemotherapy determined by interim FDG-PET in advanced-stage diffuse large B-cell lymphoma. J Clin Oncol 2010;28:1896–1903.
  • Moskowitz C, Hamlin PA, Maragulia J, et al. Sequential dose-dense RCHOP followed by ICE consolidation (MSKCC protocol 01-142) without radiotherapy for patients with primary mediastinal large B-cell lymphoma. Blood 2010;116(Suppl. 1): Abstract 420.
  • Avigdor A, Sirotkin T, Kedmi M, et al. The impact of R-VACOP-B and interim FDG-PET/CT on outcome in primary mediastinal large B cell lymphoma. Ann Hematol 2014 Mar 5. [Epub ahead of print]
  • Filippi AR, Piva C, Giunta F, et al. Radiation therapy in primary mediastinal B-cell lymphoma with positron emission tomography positivity after rituximab chemotherapy. Int J Radiat Oncol Biol Phy. 2013;87:311–316.
  • Vassilakopoulos TP, Angelopoulou MK, Rondogianni P, et al. The role of PET/CT after rituximab-CHOP (R-CHOP) in primary mediastinal large B-cell lymphoma (PMLBCL): response assessment, prognostic significance and implications for subsequent radiotherapy (RT). Haematologica 2013;98(Suppl. 1): Abstract 135.
  • Savage KJ, Yenson PR, Shenkier T, et al. The outcome of primary mediastinal large B-cell lymphoma (PMBCL) in the R-CHOP treatment era. Blood 2012;120(Suppl. 1): Abstract 303.
  • Martelli M, Ceriani L, Zucca E, et al. [18F] fluorodeoxy-glucose positron emission tomography predicts survival after chemoimmunotherapy for primary mediastinal large B-cell lymphoma: results of the International Extranodal Lymphoma Study Group IELSG-26 study. J Clin Oncol 2014 5 May. [Epub ahead of print]
  • Zinzani PL, Casadei B, Pellegrini C, et al. The role of rituximab and PET in the treatment of primary mediastinal B-cell lymphoma. Hematol Oncol 2013;31(Suppl. 1): Abstract 128.
  • Martelli M, Zucca E, Gospodarowicz M, et al. A randomized, multicentre, two-arm phase III comparative study assessing the role of mediastinal radiotherapy after rituximab-containing chemotherapy regimens to patients with newly diagnosed primary mediastinal large B-cell lymphoma (PMLBCL): the IELSG-37 study. Hematol Oncol 2013;31(Suppl. 1): Abstract 140.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.