Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Nov 1.
Published in final edited form as: Int J Antimicrob Agents. 2018 Aug 9;52(5):706–712. doi: 10.1016/j.ijantimicag.2018.08.005

Rare Mold Infections Caused by Mucorales, Lomentospora Prolificans and Fusarium, San Diego: The Role of Antifungal Combination Therapy

Jeffrey D Jenks 1,#, Sharon L Reed 1,2, Danila Seidel 3, Philipp Koehler 3, Oliver A Cornely 3,4, Sanjay R Mehta 1,*, Martin Hoenigl 1,5,*,#
PMCID: PMC6231988  NIHMSID: NIHMS1503272  PMID: 30099056

Abstract

Non-Aspergillus invasive mold infections (IMIs) are associated with devastating morbidity and mortality rates, and are increasingly diagnosed in immunocompromised hosts. The objective of this study was to describe the epidemiology and outcomes of non-Aspergillus IMIs at our university hospital in San Diego, California, United States. We performed a retrospective chart review of medical records of all patients with cultures growing non-Aspergillus molds at the Microbiology Laboratory in the Center for Academic Laboratory Medicine, Department of Pathology, University of California San Diego (UCSD) Health between mid-2014 and mid-2017 (3 year period). A total of 23 cases of non-Aspergillus IMIs were identified, including 10 cases of mucormycosis, 8 cases of lomentosporiosis, and 5 cases of fusariosis. Antifungal susceptibility testing was performed in 14 isolates and 10/11 Fusarium and Lomentospora isolates had MICs >16 μg/mL for voriconazole and/orposaconazole. Overall 180-day mortality was significantly lower among those who received combination antifungal therapy than among those who received single agent therapy [3/13 (23%) vs. 9/10 (90%); p=0.003]. In conclusion, Lomentospora prolificans (35% of non-Aspergillus IMIs), and Fusarium spp. (22%) accounted for high proportions of non-Aspergillus IMIs during the time period. Non-Aspergillus IMIs were detected in patients with various underlying diseases and associated with high mortality rates, which was significantly lower in those who received antifungal combination therapy.

Keywords: Zygomycosis, Scedosporium, Scedosporiosis, Mucormycosis, Fusarium solanii, Epidemiology, burn

1. Background

Despite recent advances in diagnosis and treatment, invasive mold infections (IMIs) are an important cause of morbidity and mortality globally, particularly in immunocompromised individuals [1]. The incidence of invasive aspergillosis (IA), the most common IMI, is 10–20 cases per 1 million population overall, with an incidence of 0.2–0.6% in the intensive care unit (ICU), 0.5–3.9% after hematopoietic stem cell transplant (HSCT), and 0.1–2.4% after solid organ transplant (SOT) [2]. Reported mortality rates from IA range from 30% to 60% at 12 weeks in patients with an underlying hematologic malignancy, HSCT, SOT or solid tumor and 41% at 12 months in SOT patients [1,2,3]. Prophylaxis against IA with newer triazoles such as posaconazole and voriconazole, particularly with induction chemotherapy for acute myeloid leukemia (AML) and in patients with graft versus host disease (GVHD), is now widely recommended and has helped decrease the morbidity and mortality from IA and increase overall survival [46].

However, the selective pressure of antifungal prophylaxis may be contributing to the emergence of less common IMIs [7]. Mucormycosis, the second most common IMI, is caused by widely prevalent fungi found in decaying organic matter and accounts for 8% of invasive fungal infections after HSCT [3] and 2% after SOT [8], with an incidence rate of 1.7 cases per 1 million population and mortality rates averaging 54%. Other filamentous fungi such as Scedosporium spp, Lomentospora spp, and Fusarium spp are also emerging opportunistic pathogens in immunocompromised individuals with incidence rates 3–8 times lower than the Mucorales. Scedosporium and Lomentospora spp are commonly found in soil and polluted waters and account for 1.6% of infections after HSCT and 0.9% of IFIs after SOT [8]. Fusarium spp are major plant pathogens and account for 3.2% of IFIs after HSCT, and 0.5% of IFIs after SOT [8]. All can cause serious, invasive infections and are associated with mortality rates between 30% and 77% for Scedosporium and Lomentospora infections [8]. Invasive fusariosis has also been associated with very high mortality rates of 79% at 90 days in patients with underlying hematologic malignancies and 87% in HSCT recipients [9] when treated with deoxycholate amphotericin B. Survival rates for invasive fusariosis have increased since the introduction of lipid formulation of amphotericin B (53% survival) and voriconazole (60% survival) [9].

The goal of this study was to investigate the risk factors, clinical manifestations, treatment modalities, and outcomes in patients with rare IMIs at our institution in San Diego, California.

2. Methods

All patients who had a non-Aspergillus mold isolated in any sample/material in the Microbiology Laboratory at University of California San Diego (UCSD) Health (San Diego, CA, USA) between July 1, 2014 and July 1, 2017 were included in the study. We then performed a retrospective chart review of medical records of all of these potential cases with non-Aspergillus mold isolates to determine if the positive cultures represented true invasive infection or colonization. Isolates were determined to represent colonization if there was either a lack of compatible findings of invasive disease on imaging, the treating physicians documented that the isolates represented colonization rather than true infection, and/or no antifungal therapy was initiated in response to these positive microbiologic findings. Conversely, isolates were determined to represent true infection if there were compatible findings on imaging and clinical findings consistent with invasive infection, and the treating physicians determined that the microbiologic findings represented true infection and antifungal therapy was initiated. Only cases in adult patients over the age of 18 were included in the analysis. Cases were classified according to revised EORTC/MSG criteria, which have been established for classifying proven IMIs in all types of cases, and probable and possible IMIs only in the subset of individuals with underlying hematologic malignancies or who received a SOT. Cases without proven infection and without underlying hematological malignancies and who were not recipients of solid organ transplantation were classified as “not classifiable”. Clinical data were compiled using the web-based registry FungiScope™ [1].

In vitro susceptibilities were determined in a total of 14 strains by a broth microdilution technique following the guidelines of the Clinical and Laboratory Standards Institute (CLSI) M38A document. Antifungal susceptibility testing was performed at the University of Texas San Antonio, Pathology, Fungus Testing Laboratory, San Antonio, Texas in 2014 and at the Associated Regional and University Pathologists, Inc. (ARUP) Laboratories, Salt Lake City, Utah, United States (2015–2017). Results were read after 48h. All azoles were tested in concentrations ranging from 0.016 to 16 μg/ml, all echinocandins and amphotericinB were tested in concentrations ranging from 0.0625 to 8 μg/ml, while terbinafine was tested in concentrations from 0.0625 to 2 μg/ml.

Statistical analyses were performed using SPSS 23 (SPSS Inc., Chicago, IL, USA). Proportions were compared using Fishers Exact test for 2 groups, and Chi-squared testing for 3 groups. A two-sided P-value of less than 0.05 was considered statistically significant. The Human Research Protections Program at the University of California, San Diego approved the study protocol and all study-related procedures (Project #171104).

3. Results

A total of 62 adult cases with non-Aspergillus mold isolates were identified over the 3-year study period, of which 23 cases had sufficient clinical data available and were determined to represent invasive infection (60% of cases with Mucor isolates, 40% of cases with Rhizopus isolates, 57% of cases with Lomentospora prolificans isolates, and 18% of cases with Fusarium isolates).

We focused our analysis on the 23 cases of invasive non-Aspergillus IMI (Table 1 and Table 2), including the 10 (43%) caused by Mucorales spp (6 by Mucor and 4 by Rhizopus; case 7 had later also detection of Trichosporon asahii; 8 proven cases, 2 probable cases), 8 (35%) by Lomentospora prolificans (case 16 had also detection of Scedosporium apiospermum in a later sputum culture, case 18 had later also detection of Mucor sp; 6 proven cases, 1 probable case and 1 not classifiable), and 5 (22%) by Fusarium spp (4 proven cases and 1 probable case). Overall, 35% of infections (8/23) occurred in patients with underlying hematologic malignancy or after SOT, while 26% (6/23) occurred in burn patients, 17% (4/23) in patients with diabetes mellitus, and 13% (3/23) after trauma or in patients in the ICU.

Table 1:

Demographic Characteristics, Underlying Diseases and Survival.

Mucormycosis
(n=10)
Lomentosporios
is
(n=8)
Fusariosis
(n=5)
Female Sex 4 5 2
Age (median, range) 47 (18–81) 53 (18–69) 45 (23–63)
Underlying Diseases/Main Risk Factors
Hematological Malignancies 3 2 1
Burn 3 - 3
Uncontrolled Diabetes 3 1 -
Lung Transplant/Cystic Fibrosis - 2 -
ICU/Polytrauma 1 2 -
Liver Disease - - 1
Chronic Granulomatous Disease - 1 -
Source of Isolate
Blood Culture - 2 -
Lung / BALF 3 2 -
Deep Soft Tissue / Biopsies 2 1 3
Eye - 3 -
Sinuses 5 - 1
Peritoneal Fluid - - 1
Survival day 180 4 4 3

ICU, intensive care unit; BALF, bronchoalveolar lavage fluid

Table 2:

Cases of non-Aspergillus Invasive Mold infections (IMI): Underlying Diseases, IMI Characteristics, Treatment and Outcome.

Cas
e
num
ber
Primar
y
Underl
ying
Diseas
e
Antifun
gals
within
14 Days
before
Diagno
sis =
Day 0
(Duratio
n in
Days)
Source
of
Isolate
IMI
Classifi
cation
Antifun
gal
Treatme
nt (Day
of
Initiatio
n)
Surgery Outcome
(final
assessment)
Surv
ival
day
180
Mucormycosis
1 Trauma
ICU
LipAmp
hB (Day
-8 – Day
0),
Micafun
gin (Day
-10 –
Day -3),
Flucona
zole
(Day -15
– Day -
11)
Soft
Tissue,
Biopsies
from:
stomach
,
omentu
m,
abdomin
al wall,
Colon/S
plenic
flexion
Proven LipAmp
hB (Day
-8) &
Posacon
azole
(Day 4;
combina
tion)
Stomac
h,
sleeve
resectio
n,
Colon/S
plenic
flexion
resectio
n
Progression/un
controlled
disease
(day 13)
No
2 Acute
Myeloid
Leuke
mia
LipAmp
hB &
Posacon
azole
(combin
ation;
Day -7 –
Day 0)
Sinuses,
Intraope
rative
Tissue
(2×)
Proven LipAmp
hB (Day
-7) &
Posacon
azole
(Day -7)
&
Micafun
gin (Day
2;
combina
tion)
Debride
ment
Complete
response
(day 330)
Yes
3 Uncont
rolled
Diabete
s
mellitus
LipAmp
hB &
Micafun
gin
(combin
ation;
Day -3 –
Day 0)
Sinuses Proven LipAmp
hB (Day
-3) &
Posacon
azole
(Day 6;
combina
tion)
- Partial
response
(day 56)
Yes
4 Burn LipAmp
hB (Day
-10 –
Day -7),
Micafun
gin (Day
-10 –
Day 0)
Soft
Tissue
Proven LipAmp
hB &
Posacon
azole
(Day 0;
combina
tion)
Debride
ment
Complete
response
(day 42)
Yes
5 Uncont
rolled
Diabete
s
mellitus
(ICU)
Flucona
zole
(Day -4
– Day 0)
BALF,
Sputum,
Lung
Tissue
Proven Micafun
gin (Day
-2)
- Progression/un
controlled
disease (day 2)
No
6 Uncont
rolled
Diabete
s
mellitus
- Sinuses,
Hard
palate
biopsy
Proven LipAmp
hB &
Posacon
azole
(Day 0;
combina
tion)
Debride
ment
Progression/un
controlled
disease
(day 22)
No
7 Acute
Lymph
atic
Leuke
mia
Posacon
azole
(Day -44
– Day 0)
BALF Probable LipAmp
hB (Day
0)
- Progression/un
controlled
disease
(day 15)
No
8 Burn Voricon
azole
(Day -30
– Day 0)
Sinuses
(6×)
Proven LipAmp
hB (Day
0)
Debride
ment
Progression/un
controlled
disease
(day 26)
No
9 Acute
Lymph
atic
Leuke
mia
Posacon
azole
(Day -17
– Day 0)
BALF Probable LipAmp
hB (Day
0)
- Progression/un
controlled
disease
(day 17)
No
10 Burn Voricon
azole
(Day -3
– Day
10),
Flucona
zole
(Day -12
– Day 0)
Sinuses
(5×)
Proven LipAmp
hB (Day
0) &
Posacon
azole
(Day 10;
combina
tion)
Debride
ment
Complete
response
(day 104)
Yes
Lomentosporiosis
11 Uncont
rolled
Diabete
s
Mellitus
NA Eye Proven Voricon
azole
systemic
&
intravitre
al (Day
0)
Right
Eye
Enuclea
tion
Progression/un
controlled
disease (day 3)
No
12 Chronic
Cardio-
vascula
r
Diseas
e (ICU)
Flucona
zole
(Day -4
– Day -
2)
Eye (2×) Proven Voricon
azole
(Day -1)
&
Terbinafi
ne (Day
0;
combina
tion) +/−
Micafun
gin (Day
2 – Day
9)
Left Eye
Enuclea
tion
Partial
Response
(day 75)
Yes
13 Non
Hodgki
n
Lymph
oma
Micafun
gin (Day
-11 –
Day 0),
Flucona
zole
(Day -11
– Day -
9)
Blood
Culture
(2×)
Proven Micafun
gin (Day
-11),
LipAmp
hB (Day
5)
- Progression/un
controlled
disease (day 6)
No
14 Multiple
Myelo
ma
Lip
AmphB
intravitre
al (Day -
5)
Eye
(twice)
Proven Lip
AmphB
systemic
and
intravitre
al (Day
0)
Left Eye
Vitrecto
my
Progression/un
controlled
disease (day 7)
No
15 Lung
Transpl
ant
Recipie
nt (4
years
ago);
Cystic
Fibrosi
s
Posacon
azole
(Day -31
– Day 0)
BALF Probable Voricon
azole &
Micafun
gin &
Terbinafi
ne (Day
2;
combina
tion)
- Stable disease
(day 84)
Yes
16 Cystic
Fibrosi
s
NA Sputum
Not
classifia
ble
Voricon
azole &
Micafun
gin (Day
0;
combina
tion)
- Stable disease
(day 84)
Yes
17 Chronic
granulo
-
matous
disease
Micafun
gin (Day
-12 –
Day -8),
Flucona
zole
(Day -31
– Day 0)
Blood
culture
Proven Voricon
azole
(Day 0)
&
Terbinafi
ne (Day
2;
combina
tion)
- Complete
response (day
42)
Yes
18 Major
Surger
y (ICU)
Micafun
gin &
LipAmp
hB (Day
-15 –
Day 0;
combina
tion)
Deep
soft
tissue
(7×)
Proven Voricon
azole &
LipAmp
hB (Day
0;
combina
tion),
then
Posacon
azole &
Terbinafi
ne (Day
21;
combina
tion)
Debride
ment
Stable Disease
(day 115)
No
Fusariosis
19 Chronic
lympho
cytic
leukemi
a
NA Sinuses
(5×)
Proven Voricon
azole
(Day 0)
&
Terbinafi
ne (Day
4;
combina
tion)
Debride
ment of
sinuses
Stable disease
(day 230)
Yes
20 Burn Micafun
gin (Day
-3 – Day
0),
Flucona
zole
(Day -12
– Day -
5)
Skin/soft
tissue
(2×)
Proven LipAmp
hB (Day
-3) &
Voricon
azole
(Day 0;
combina
tion)
Debride
ment
Complete
response
(day 42)
Yes
21 Alcohol
ic liver
disease
Micafun
gin (Day
-20 –
Day 0),
Flucona
zole
(Day -6
- Day -
3)
Peritone
al fluid
Proven LipAmp
hB (Day
0)
- Progression/un
controlled
disease (day 2)
No
22 Burn Flucona
zole
(Day -16
– Day 0)
Skin/soft
tissue
(2×)
Proven LipAmp
hB (Day
0)
Debride
ment
Stable disease
(day 54)
No
23 Burn NA Skin/soft
tissue
(8×)
Sterile
fluid (×)
Proven Voricon
azole
(Day 0)
Debride
ment
Complete
response
(day 183)
Yes

Abbreviations: BALF, bronchoalveolar lavage fluid; ICU, intensive care unit; LipAmphB, liposomal Amphotericin B

Table 1 shows demographic characteristics, underlying diseases, source of isolates and survival for each group of IMI. No significant difference was observed in underlying diseases between the three groups (p=0.142), while significant differences were observed regarding the source of the fungal isolate (p=0.017), with Mucorales being more frequently isolated from sinuses and Lomentospora prolificans being more frequently isolated from eyes.

Table 2 shows patient and disease characteristics as well as treatment and outcome for all 23 cases. Overall 180-day mortality was 52% (12/23), and significantly lower among those who received combination antifungal therapy than among those who received single agent therapy [3/13 (23%) mortality among those with combination therapy vs. 9/10 (90%) mortality among those with single agent therapy; p=0.003].

Out of 10 cases of mucormycosis (4 caused by Rhizopus spp. and 6 by Mucor spp.), 6 died within 30 days of detection of Mucorales; all 4 survivors received combination therapy with liposomal amphotericin B and posaconazole, while only 2/6 non-survivors received combination therapy (p=0.076).

Table 3 shows results of antifungal susceptibility testing and minimum inhibitory concentrations (MIC). Antifungal susceptibility testing revealed that 10/11 Lomentospora prolificans and Fusarium spp isolates had MICs >16 μg/mL against voriconazole and/or posaconazole. Among cases with Lomentospora prolificans infections, all four survivors received combination therapy with either voriconazole plus terbinafine (n=3) or voriconazole plus micafungin (n=1), while 1/4 non-survivors received also combination therapy (p=0.143). In patients with invasive fusariosis, treatment with voriconazole alone or in combination showed a trend to being associated with survival (3/3 survived, while both patients who did not receive voriconazole did not survive; p=0.100).

Table 3.

Results of Antifungal Susceptibility Testing (performed in 14/23 isolates). Minimum inhibitory concentrations (MIC) displayed.

Case
number
Isolate AF Before
Isolation
MIC (mg/L)
2 Rhizopus sp. LipAmphB &
Posaconazole
(combination)
LipAmphB: 1
Itraconazole: 1
Posaconazole: 0.5
Voriconazole: 8
3 Rhizopus sp. LipAmphB,
Micafungin
(combination)
LipAmphB: 2
Itraconazole: 2
Posaconazole: 1
Voriconazole: >16
10 Mucor sp. Voriconazole,
Fluconazole
LipAmphB: 0.5
Itraconazole: >16
Voriconazole: >16
11 Lomentospora
prolificans
NA LipAmphB: >8
Itraconazole: >16
Posaconazole: >16
Voriconazole: >16
Anidulafungin: 4
Caspofungin: >8
Micafungin: >8
12 Lomentospora
prolificans
Fluconazole Posaconazole: >16
Anidulafungin: 1
14 Lomentospora
prolificans
Lip AmphB
systemic and
intravitreal
Posaconazole: >16
Terbinafine: >2
15 Lomentospora
prolificans
Posaconazole LipAmphB: >8
Itraconazole: >16
Posaconazole: >16
Voriconazole: >16
Anidulafungin: >8
Caspofungin: >8
Micafungin: 1
16 Lomentospora
prolificans
NA Itraconazole: >16
Posaconazole: 1
Anidulafungin: 2
Caspofungin: 1
Micafungin: 0.25
17 Lomentospora
prolificans
Micafungin,
Fluconazole
LipAmphB: >8
Posaconazole: >16
Voriconazole: >16
Anidulafungin: <0.0625
Caspofungin: <0.0625
Micafungin: <0.0625
Terbinafine: 2
18 Lomentospora
prolificans
Micafungin &
LipAmphB
(combination)
Posaconazole: >16
Anidulafungin: >8
Caspofungin: >8
Micafungin: >8
19 Fusarium
solanii
NA LipAmphB: 2
Posaconazole: >16
Voriconazole: 16
Caspofungin: >8
Isavuconazole: >16
Terbinafine: 0.25
20 Fusarium sp. Micafungin,
Fluconazole
LipAmphB: 2
Itraconazole: >16
Posaconazole: >16
Voriconazole: >16
21 Fusarium sp. Micafungin,
Fluconazole
LipAmphB: 2
Itraconazole: >16
Voriconazole: >16
22 Fusarium sp. Fluconazole LipAmphB: >8
Itraconazole: >16
Voriconazole: >16

Abbreviations: AF, antifungals; LipAmphB, liposomal Amphotericin B; NA, not applicable

4. Discussion

Invasive infection due to non-Aspergillus molds is an important cause of morbidity and mortality, particularly in immunocompromised individuals. In this study, infections occurred in patients with a variety of underlying diseases and diverse sites, with mucormycosis most likely isolated from the sinuses, Lomentospora prolificans from the eye, and Fusarium from soft tissue. Thus, invasive infection from these molds can occur in individuals without classically-defined immunocompromising drugs and conditions, (e.g., HSCT or SOT), and can occur in a variety of sites. Clinicians should be observant for signs of these infections in the right clinical context. Overall, non-Aspergillus IMIs were associated with high mortality rates, particularly in cases with single agent antifungal therapy (9/10 died, 90%), while mortality was significantly lower in those who received combination antifungal therapy (2/13 died, 23%).

High mortality rates from non-Aspergillus molds were noted in this study, similar to previous studies [3, 8, 9]. Mortality at 180 days ranged from 40% with invasive fusariosis, 50% with Lomentospora infection, and 60% with mucormycosis. There was an association between survival and the use of combination therapy, driven in particular by patients with mucormycosis and Lomentospora infections, with a trend towards improved survival in both. Of the 10 patients with mucormycosis, 6 received combination therapy with liposomal amphotericin B plus posaconazole, with one patient receiving treatment with micafungin as well. Of those who received combination therapy, 4/6 survived, with none surviving in those that received monotherapy. The European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and the European Conference on Infections in Leukemia (ECIL-6) guidelines recommend liposomal amphotericin B as first-line therapy (AII and BII recommendations, respectively) for the management of invasive mucormycosis [10], although posaconazole has shown good efficacy for salvage treatment of mucormycosis [11].

There is some data supporting combination therapy for the treatment of infections from mucormycosis. In-vitro studies with combination of amphotericin B and posaconazole has demonstrated synergy against Rhizopus isolates [12]. Combination therapy with amphotericin B and posaconazole in animal models has yielded mixed results. In one study investigating combination therapy with amphotericin B plus posaconazole versus monotherapy with amphotericin B in diabetic ketoacidotic or neutropenic mice with disseminated mucormycosis, combination therapy did not result in improved survival [13]. However, in another study in immunosuppressed mice, amphotericin B plus posaconazole improved survival and reduced fungal tissue burden compared to monotherapy with either drug in mice with disseminated mucormycosis [14].

In terms of clinical data, a retrospective study of diabetic patients with rhino-orbital or rhino-orbital-cerebral mucormycosis showed that combination therapy with amphotericin B and caspofungin was associated with greater 30-day survival compared to monotherapy with amphotericin B (100% versus 45%), although the sample size was small [15]. Another retrospective study examined combination therapy with amphotericin B and posaconazole to treat invasive mucormycosis in 32 patients with hematologic malignancy or aplastic anemia [16]. Most patients initially received monotherapy with amphotericin B, with posaconazole added as salvage therapy due to lack of response with amphotericin B alone. At 3 months, those patients who received both antifungal agents did not have worse survival, although posaconazole was used as salvage rather than combination therapy [16]. Another large retrospective study of 106 patients with underlying hematologic malignancy or HSCT recipients with mucormycosis investigated outcomes between patients treated with monotherapy and combination therapy at a single medical center from 1994 to 2014. This study did not find an overall mortality benefit between those treated with monotherapy and combination therapy at 6-weeks (43% versus 41%, respectively), although those receiving combination therapy with amphotericin B plus posaconazole had a higher rate of survival compared to those receiving monotherapy (24/32 survived versus 27/47, respectively) [17]. Thus, combination therapy with liposomal amphotericin B with posaconazole may be more efficacious than monotherapy with amphotericin B, although further investigation is warranted.

In line with previous studies, high MICs against most antifungals were observed for Lomentospora prolificans isolates, with some isolates displaying lower MICs for echinocandins and one isolate displaying a low MIC for posaconazole. Of the 8 patients with Lomentospora infections, 5 received combination therapy with voriconazole plus at least one other agent (in 4/5 patients the combination included voriconazole and terbinafine). Of those receiving combination therapy 80% (4/5) survived, while no patients who received monotherapy survived. Combination treatment (primarily broad spectrum azole plus terbinafine) is also the recommended treatment (BII recommendation) for the treatment of Lomentospora prolificans infections by the ESCMID and the European Confederation of Medical Mycology (ECMM) [18]. This recommendation is mostly based on case reports demonstrating clinical efficacy with combination voriconazole and terbinafine, while data from large scale studies to support this approach is lacking given the rareness of these infections.

Notably, the majority of Fusarium isolates were resistant to both first-line and salvage therapy. Of the 5 patients with Fusarium infection, 4 had antifungal susceptibility testing; of these, all 4 had an MIC ≥16 to voriconazole, and of those tested against posaconazole (2/4), both had a MIC >16 mg/L. In other studies the MIC of voriconazole and posaconazole against Fusarium ranged from 1.0 – 16.0 mg/L and 0.25 – 32 mg/L [19], respectively. Nevertheless, studies have shown the benefit of voriconazole-based treatment regimens for survival of invasive fusariosis [9], and a similar trend was also observed in our study (all patients with voriconazole based treatment regimens survived, while both patients who did not receive voriconazole did not survive). However, this difference may also be explained by the fact that all survivors received surgery, which plays a major adjuvant role in the treatment of these infections, particularly when high MICs are noted, as in this study.

This non-randomized study does have several limitations and our main finding that combination therapy was strongly associated with a better outcome should therefore be interpreted with caution. This is a retrospective cohort study done at a single medical institution in San Diego, so these findings may not be representative of other patient populations. Still, the patients in this study had a wide variety of predisposing factors increasing their risk for IMI, resulting in a diverse cohort. In addition, the sample size was low, although this is a natural limitation of studies looking at rare diseases such as those documented here. This study was mostly descriptive in character and underpowered to assess for clear associations, such as antifungal treatment and survival, for example. Finally, none of the cases received isavuconazole, which has recently been shown to be a promising therapeutic option for non-Aspergillus IMIs and also IMIs caused by more than one fungal species [20]. Nevertheless, this study adds to the current body of literature investigating rare IMIs.

5. Conclusions

In conclusion, this study describes non-Aspergillus IMIs in patients with various underlying diseases that resulted in high mortality rates. Notably, of these IMIs Lomentospora prolificans (35%) and Fusarium spp. (22%) were emerging pathogens, with the vast majority of isolates resistant to both voriconazole and posaconazole, the two agents preferred for the treatment of these infections. Overall, mortality rates were significantly lower in patients who received antifungal combination therapy. Further investigation is needed to determine the optimal treatment for these infections, including if combination antifungal therapy offers a survival benefit over monotherapy.

Highlights.

  • Retrospective analysis of patients diagnosed with non-Aspergillus invasive mold infections at the University of California San Diego Medical Center, San Diego, California, United States.

  • IMIs occurred in patients with a variety of underlying diseases and diverse sites, not just those with classically-defined immunocompromising drugs and conditions

  • Most Fusarium and Lomentospora isolates had MICs >16 μg/mL for voriconazole and/or posaconazole

  • Overall 180-day mortality was significantly lower among those who received combination antifungal therapy [3/13 (23%)] than among those who received single agent therapy [9/10 (90%); p=0.003]

Acknowledgments

Declarations

Funding: This work was supported by Grants from the National Institutes of Health (AI036214, MH113477, and AI106039). The Fungiscope database, which was used for data collection, is supported by unrestricted grants from Astellas Pharma, Gilead Sciences, MSD/Merck, and Pfizer Pharma GmbH. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The funders had no role in study design, data collection, analysis, interpretation, decision to publish, in the writing of the manuscript, and in the decision to submit the manuscript for publication.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Competing Interests: OAC is supported by the German Federal Ministry of Research and Education and the European Commission, and has received research grants from, is an advisor to, or received lecture honoraria from Actelion, Amplyx, Arsanis, Astellas, AstraZeneca, Basilea, Cidara, Da Volterra, Duke University (NIH UM1AI104681), F2G, Gilead, GSK, Janssen, Leeds University, Matinas, Medicines Company, MedPace, Menarini, Merck/MSD, Miltenyi, Paratek, Pfizer, PSI, Rempex, Roche, Sanofi Pasteur, Scynexis, Seres, Summit, Tetraphase, and Vical. PK reports non-financial support from Merck, non-financial support from MedImmune and lecture honoraria from Astellas. MH received research funding from Gilead and reports lecture honoraria from the Research Practitioner Network, Gilead, Basilea and Merck. All other authors have none to declare.

Ethical Approval: The Human Research Protections Program at the University of California, San Diego approved the study protocol and all study-related procedures (Project #171104).

References

  • 1.Cornely OA, Lass-Flörl C, Lagrou K, Arsic-Arsenijevic V, Hoenigl M. Improving outcome of fungal disease – Guiding experts and patients towards excellence. Mycoses 2017. July;60(7):420–425. [DOI] [PubMed] [Google Scholar]
  • 2.Morgan J, Wannemuehler KA, Marr KA, Hadley S, Kontoyiannis DP, Walsh TJ, et al. Incidence of invasive aspergillosis following hematopoietic stem cell and solid organ transplantation: interim results of a prospective multicenter surveillance program. Med Mycol 2005. May;43 Suppl 1:S49–58. [DOI] [PubMed] [Google Scholar]
  • 3.Kontoyiannis DP, Marr KA, Alexander BD, Anaissie EJ, Walsh TJ, Ito J, et al. Prospective surveillance for invasive fungal infections in hematopoietic stem cell transplant recipients, 2001–2006: overview of the Transplant-Associated Infection Surveillance Network (TRANSNET) Database. Clin Infect Dis 2010. April 15;50(8):1091–100. [DOI] [PubMed] [Google Scholar]
  • 4.Ullmann AJ, Aguado JM, Arikan-Akdagli S, Denning DW, Groll AH, et al. Diagnosis and management of Aspergillus diseases: executive summary of the 2017 ESCMID-ECMM-ERS guideline. Clin Microbiol Infect 2018. March 12 Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 5.Patterson TF, Thompson GR, Denning DW, Fishman JA, Hadley S, et al. Practice guidelines for the diagnosis and management of aspergillosis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis 2016. August 15;63(4):e1–e60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Eigl S, Hoenigl M, Spiess B, Heldt S, Prattes J, Neumeister P, et al. Galactomannan testing and Aspergillus PCR in same-day bronchoalveolar lavage and blood samples for diagnosis of invasive aspergillosis. Med Mycol 2017. July 1;55(5):528–534. [DOI] [PubMed] [Google Scholar]
  • 7.Auberger J, Lass-Flörl C, Aigner M, Clausen J, Castl G, Nachbaur D. Invasive fungal breakthrough infections, fungal colonization and emergence of resistant strains in high-risk patients receiving antifungal prophylaxis with posaconazole: real-life data from a single-centre institutional retrospective observational study. J Antimicrob Chemother 2012. September;67(9):2268–73. [DOI] [PubMed] [Google Scholar]
  • 8.Park BJ, Pappas PG, Wannemuehler KA, Alexander BD, Anaissie EJ, Andes DR, et al. Invasive non-Aspergillus mold infections in transplant recipients, United States, 2001–2006. Emerg Infect Dis 2011. October;17(10):1855–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nucci M, Marr KA, Vehreschild MJ, de Souza CA, Velasco E, Cappellano P, et al. Improvement in the outcome of invasive fusariosis in the last decade. Clin Microbiol Infect 2014. June;20(6):580–5. [DOI] [PubMed] [Google Scholar]
  • 10.Cornely OA, Arikan-Akdagli S, Dannaoui E, Groll AH, Lagrou K, Chakrabarti A, et al. ESCMID and ECMM joint guidelines for the diagnosis and management of mucormycosis 2013. Clin Microbiol Infect 2014. April;20 Suppl 3:5–26. [DOI] [PubMed] [Google Scholar]
  • 11.van Burik JA, Hare RS, Solomon HF, Corrado ML, Kontoyiannis DP. Posaconazole is effective as salvage therapy in zygomycosis: a retrospective summary of 91 cases. Clin Infect Dis 2006. April 1;42(7): e61–5. [DOI] [PubMed] [Google Scholar]
  • 12.Perkhofer S, Locher M, Cuenca-Estrella M, Ruchel R, Wurzner R, Dierich MP, et al. Posaconazole enhances the activity of amphotericin B against hyphae of zygomycetes in vitro. Antimicrob Agents Chemother 2008. July;52(7):2636–2638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ibrahim AS, Gebremariam T, Schwartz JA, Edwards JE Jr, Spellberg B. Posaconazole mono- or combination therapy for treatment of murine zygomycosis. Antimicrob Agents Chemother 2009. February;53(2):772–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rodriguez MM, Serena C, Marine M, Pastor FJ, Guarro J. Posaconazole combined with amphotericin B, an effective therapy for a murine disseminated infection caused by Rhizopus oryzae. Antimicrob Agents Chemother 2008. October;52(10:3786–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Reed C, Bryant R, Ibrahim AS, Edwards J Jr, Filler SG, Goldberg R, et al. Combination polyene-caspofungin treatment of rhino-orbital-cerebral mucormycosis. Clin Infect Dis 2008. August 1;47(3):364–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Pagano L, Cornely OA, Busca A, Caira M, Cesaro S, Gasbarrino C, et al. Combined antifungal approach for the treatment of invasive mucormycosis in patients with hematologic disease: a report from the SEIFEM and FUNGISCOPE registries. Haematologica 2013. October;98(10):e127–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kyvernitakis A, Torres HA, Jiang Y, Chamilos G, Lewis RE, Kontoyiannis DP. Initial use of combination treatment does not impact survivial of 106 patients with haematologic malignancies and mucormycosis: a propensity score analysis. Clin Microbiol Infect 2016. September;22(9):811.e1–811e8. [DOI] [PubMed] [Google Scholar]
  • 18.Tortorano AM, Richardson M, Roilides E, van Diepeningen A, Caira M, Munoz P, et al. ESCMID and ECMM joint guidelines on diagnosis and management of hyalohyphomycosis: Fusarium spp., Scedosporium spp. and others. Clin Microbiol Infect 2014. April; 20 Suppl 3:27–46. [DOI] [PubMed] [Google Scholar]
  • 19.Guinea J, Pelaez T, Recio S, Torres-Narbona M, Bouza E. In vitro antifungal activities of isavuconazole (BAL4815), voriconazole, and fluconazole against 1,007 isolates of Zygomycete, Candida, Aspergillus, Fusarium, and Scedosporium species. Antimicrob Agents Chemother 2008. April;52(4):1396–1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jenks JD, Salzer HJ, Prattes J, Krause R, Buchheidt D, Hoenigl M. Spotlight on isavuconazole in the treatment of invasive aspergillosis and mucormycosis: design, development, and place in therapy. Drug Des Devel Ther 2018. April 30;12:1033–1044. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES