All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit the International Myeloma Foundation or HealthTree for Multiple Myeloma.

The Multiple Myeloma Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your Multiple Myeloma Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The Multiple Myeloma Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the Multiple Myeloma Hub cannot guarantee the accuracy of translated content. The Multiple Myeloma Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2020-05-19T13:54:45.000Z

Is t(14;16) an independent prognostic factor in patients with multiple myeloma?

May 19, 2020
Share:

Bookmark this article

Translocations of the immunoglobulin heavy-chain gene (IgH) locus at 14q32 are involved at early stages of multiple myeloma (MM) pathogenesis and occur in approximately 40% of cases. Different mutations confer different risk on prognosis: t(11;14), the most common lesion, is considered a standard-risk chromosomal abnormality (CA); while t(4;14), t(14;16), and t(14;20) have been historically associated with higher-risk disease.

Since the publication of the Revised International Staging System (R-ISS) in 2015 by Palumbo et al.1, the t(14;16) was incorporated to the clinical practice as a CA associated with poor prognosis in patients newly diagnosed with MM (NDMM). However, multiple studies have been published questioning the direct impact of t(14;16) on the survival of these patients.

The main challenge when studying t(14;16) is that its incidence is relatively rare: it is only observed in 3–5% patients with MM. Therefore, all previous analyses included a limited number of patients with t(14;16) and were probably underpowered to define its prognostic value. To further elucidate the clinical effect of t(14;16) in a context where multiple new drugs have been incorporated to the MM therapeutic arsenal, Mina et al.2 recently published one of the largest studies to date in Blood Cancer Journal, which included data from 123 patients with t(14;16) confirmed by centralized fluorescent in situ hybridization (FISH) at diagnosis.

The authors selected patients diagnosed between 2006–2017 with symptomatic MM and t(14;16)-positive FISH, registered in multiple databases from different European and American centers. When analyzing the patient characteristics at diagnosis (Table 1), one of the findings consistent with previous studies is that t(14;16) is rarely found as a single mutation (only in ten patients in this cohort). Interestingly, most patients presented concomitant CAs such as del(13q) (71%), amp(1q) (51%), and del(17p) (23%), all considered to be associated with a worse prognosis.

Table 1. Patient characteristics at diagnosis and first-line treatment2

ASCT, autologous stem cell transplantation; FISH, fluorescent in situ hybridization; IMiD, immunomodulatory drug; ISS, International Staging System; PI, proteasome inhibitor; ULN, upper limit of normal

Characteristics

Patients (N = 123)

Age, median (range)

66 (38–87)

Bone marrow plasma cells (%), median (range)

60 (0–100)

Lactate dehydrogenase, > ULN (%)

23 (23)

ISS, n (%)

1
2
3
Missing

 

25 (20)
41 (34)
53 (43)
4 (3)

FISH, n (%)

del(13q)

amp(1q)

del(17p)

t(4;14)

t(11;14)

 

87 (71)

33 (51)

27 (23)

14 (11)

6 (6)

Induction treatment, n (%)

IMiD®-based (thalidomide or lenalidomide)
PI-based (bortezomib)
IMiD plus PI-based

 

52 (42)
36 (29)
34 (28)

ASCT, n (%)

52 (42)

Maintenance, n (%)

50 (41)

After a median follow-up of 53 months (95% CI, 35–63), the median progression-free survival (PFS) was 19 months (95% CI, 16-30) and overall survival (OS) was 53 months (95% CI, 36-63) for the full cohort of patients with t(14;16). At 5 years, 39% were still alive. When analyzing the results according to cytogenetics, patients with additional CAs presented a significantly shorter PFS and shorter OS, compared to those without them (Table 2).

If the concomitant CA was del(13q), the risk of progression or death increased by 1.64-fold (p = 0.05), and OS was significantly shortened (HR, 1.96; p = 0.04). If the concomitant CA was del(17p), the risk of progression or death increased by 1.35-fold (p = 0.23) and this was 2.2-fold (p = 0.02) in patients with amp(1q). The significance of these comparisons, though, needs to be evaluated carefully since sample size of patients with no concomitant CAs was small.

Table 2. Outcomes based on CAs2

CAs, chromosomal abnormalities; HR, hazard ratio; OS, overall survival; PFS, progression-free survival

Outcomes

Patients with t(14;16) and additional CAs (N = 101)

Patients with t(14;16) only (N = 10)

HR

p value

PFS, months

17

NR

3.33

0.04

OS, months

46

NR

1.54

0.47

In addition to concomitant CAs, other characteristics negatively influenced the outcomes of patients with t(14;16) in the multivariate analysis. A shorter PFS was observed in patients classified as ISS-3, and hypercalcemia, elevated LDH, and again, ISS-3, were all correlated with a significantly shorter OS.

Lastly, in patients who were eligible for autologous stem cell transplantation (ASCT), the upfront transplant prolonged the median PFS compared to standard chemotherapy without ASCT, as did the use of maintenance therapy. Another factor associated with prolonged PFS was the achievement of complete response (CR), or better, as best response:

  • ASCT vs no ASCT: 36 vs 17 months (HR, 0.5; p = 0.036)
  • Maintenance vs no maintenance: 36 vs 19 months (HR, 0.59; p = 0.045)
  • ≥ CR vs partial response (PR) or very good PR (HR, 0.29; p < 0.001)

In summary, according to this analysis, almost every patient with t(14;16) presented with at least one other high-risk characteristic at diagnosis, which are associated with poorer survival outcomes.

These results are in line with those reported recently by Goldman-Mazur et al.,3 in the only other study with an equally high sample size of patients with t(14;16) (N = 223). In this multicenter retrospective study, Goldman-Mazur et al. found that approximately a quarter of patients with t(14;16) initially present with renal failure. After a median follow-up of 4.1 years (95% CI, 3.7–18.7), the median PFS and OS were 2.1 years (95% CI, 1.5–2.4) and 4.1 years (95% CI, 3.3–5.5), respectively. However, the OS significantly decreased in older patients (age > 60 years) and patients with R-ISS 3 disease compared to the rest of the patients included in the study. It should be noted that in this study, as well as in Mina et al.,2 results were not compared to a matched control group of patients without t(14;16).

The question remains then, whether it is possible to link this unfavorable prognosis solely to the presence of t(14;16) or not. Moreover, the decision to include t(14;16) in the R-ISS was based on the International Myeloma Working Group (IMWG) molecular classification,4 which initially used the data from only two small studies. Although its significance was validated later in larger cohorts, t(14;16) was always analyzed along with other CAs.

On the other hand, incorporating novel agents into first-line of therapy, and using some clinical protocols specifically for patients with a high-risk profile (i.e., double ASCT), it might be possible to improve the outcomes for patients with t(14;16), masking the previously described negative effect of this CA.

Taking everything into account, is it safe to say that there is a growing need for a revision of the Revised-ISS to address how to improve the evaluation of patients diagnosed with MM in the era of novel agents, according to their cytogenic profile.

  1. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised international staging system for multiple myeloma: a report from the International Myeloma Working Group. J Clin Oncol. 2015;33(26):2863-2869. DOI: 1200/JCO.2015.61.2267

  2. Mina R, Joseph NS, Gay F, et al. Clinical features and survival of multiple myeloma patients harboring t(14;16) in the era of novel agents. Blood Cancer J. 2020;10(4):40. DOI: 10.1038/s41408-020-0307-4

  3. Goldman-Mazur S, Jurczyszyn A, Castillo JJ, et al. A multicenter retrospective study of 223 patients with t(14;16) in multiple myeloma. Am J Hematol. 2020;95(5):503-509. DOI: 10.1002/ajh.25758

  4. Fonseca R, Bergsagel PL, Drach J, et al. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia. 2009;23(12):2210-2221. DOI: 1038/leu.2009.174

Newsletter

Subscribe to get the best content related to multiple myeloma delivered to your inbox