Expired activity
Please go to the PowerPak homepage and select a course.

INVASIVE MYCOSES IN HEMATOLOGIC MALIGNANCIES: OPTIMAL MANAGEMENT STRATEGIES

SECTION 1A: INTRODUCTION

Matthew Lunning, DO

Welcome to Invasive Mycoses in Hematologic Malignancies: Optimal Management Strategies. I’m Matthew Lunning. I'm an assistant professor in the Division of Oncology and Hematology at the University of Nebraska Medical Center. By training, I am a lymphoma/myeloma transplanter. My co-panelist, Dr. Alison Freifeld, will help me moderate this session. She is a professor of internal medicine, also at the Fred & Pamela Buffett Cancer Center at the University of Nebraska Medical Center in Omaha, Nebraska.


So here's the agenda. This is the introduction component, and Dr. Freifeld will present a little bit of background in invasive fungal infections. We’ll then turn it over to Dr Maertens and Jim Lewis about invasive fungal infections diagnosis and therapeutics. And we will get started now with the symposium portion with Dr. Freifeld discussing the epidemiology of invasive fungal infections in hematologic malignancies.

SECTION 1B: EPIDEMIOLOGY OF INVASIVE FUNGAL INFECTIONS IN HEMATOLOGIC MALIGNANCIES

Alison G. Freifeld, MD

Good afternoon, and welcome to the Invasive Fungal Infection Symposium. I'm Alison Freifeld, colleague of Matt Lunning. We work together on a daily basis at the Fred & Pamela Buffett Cancer Center in Omaha.

So, as you know, infectious complications have improved in terms of management over the last 60 years that we've been using cytotoxic chemotherapy. It’s really gratifying to see that we've been able to predict and manage – and in many cases, contain – things like cytomegalovirus, and many of the routine bacterial infections. Of course, now we're faced with the rising incidence of resistance among many infectious pathogens. And today we are talking specifically about invasive fungal infections or IFI, especially in hematologic malignancies.


And so, what I’d like to do is first orient you to the epidemiology of IFIs in both hematologic malignancies and transplant. There are a number of opportunities for invasive fungal infections to occur during the course of treatment for hematologic malignancies.

And for the most part, we're talking about Aspergillus species, which typically account for about 60% of all the IFIs documented. Which are far greater in number than Candida infections, accounting for about 30%. Then, occasionally, we see zygomycoses or Fusarium in a small percentage, but they are, as you know, very difficult to treat.

if we looked to the course of a patient, starting with the AML population, data have shown us from large population studies performed prior to the advent of the triazoles that we're going to talk about today – that there was a baseline rate of about 8% to 12% of invasive fungal infections in that population, comprising all of these genera: Aspergillus, Candida, and Fusarium and the fungi of the Zygomycota phylum.

Interestingly, in the ALL population the rates are a little lower, about 6.5%. In a study performed in Italy in the early part of this century, looking at about 150 infectious aspergillosis patients in newly diagnosed AML, about 60% were diagnosed during induction, about 36% during relapse and refractory disease, as you’d expect. So most episodes of IFI occur during induction for AML. And only about 3% during consolidation cycles.

If we look at the stem-cell transplant population, we know that the vast majority of IFIs (99%) occur in the allogeneic setting as opposed to the autologous transplant group. In allogeneics, the cumulative incidence is 2 to 8%, in general. There is a very wide variety of cumulative incidence rates, depending on which population studies are looked at. But a common finding is that 80% of these typically occur within a hundred days of transplant, during the neutropenic period and also during periods of acute GvHD.


So before we really go through a lot of the data that my colleagues are going to present, we need to kind of review the definitions of IFI. And these are definitions that were created by the combined efforts of the EORTC and the Mycoses Study Group in the United States, just to be able to clarify what we're all talking about.

And the proven definition is very easy – because it requires histology, cytology, or culture data from a sterile site. So nobody would argue with that and I've shown at the right, some lung histology, H&E section demonstrating those narrow septate hyphae with acute-angle branching and tissue – very characteristic of aspergillosis. That’s the kind of data that really is required for a proven infection. And it’s unfortunately, the kind of data that we rarely are able to achieve.

A probable definition was created so that we could capture IFIs from the vast majority of patients where host factors, clinical criteria, and mycological criteria are required in order to categorize the infection as probable. The host factors have to be approximate and show severe immunosuppression, neutropenia, recent allogeneic transplant, corticosteroids, T cell immunosuppression, etc.

The clinical criteria are largely dependent on the CT appearance of what is thought to be characteristic of an invasive fungal infection, typically a mold. And in this situation, I’ve shown you a large nodular lesion on a CT scan, that I think most of us in the right setting and the right host would consider likely to be due to an invasive mold infection.

These mycologic criteria were first delineated in 2002, and then revised in 2008. They were revised to include not just mold by cytology or direct microscopy or culture in a non-sterile specimen, but also to include the new antigen testing methods.

Well, these are not necessarily brand new in 2015 – and we’ll hear more about them from Dr. Maertens. But a positive galactomannan test from serum, BAL, or CSF, or a β-D-glucan test from serum are considered enough evidence at the mycologic level to designate a probable infection.


So let's turn first to the incidence of IFI in the hematologic malignancy group—that is, the non-transplant populations. And I've accumulated here several very large studies that were done in the early part of the century to show you the splay of data in IFIs in the baseline group, meaning those who received either placebo or fluconazole prophylaxis.

There are 2 retrospective studies and 3 randomized controlled trials. And I think these will provide to us the best data, in which the control groups had anywhere between a 2% and 8% incidence of IFI, with the application of a mold-active agent, itraconazole, as prophylaxis in 2 of these studies.

I want to particularly draw your attention to this study in 2007 by Cornely, which really is kind of the basis for most studies going forward, most information going forward about the azoles and their efficacy for prophylaxis. And in this large study, many of you may be familiar with – 602 patients with AML were randomized to receive either fluconazole or itraconazole, which were the standards of care at the time versus posaconazole – in the oral suspension form, very different from the tablet form that we have today. And they recognized a dramatic decrease from 8% to 2% in the prophylaxed group with a concomitant decrease in the defined invasive aspergillosis group from 7% to 1%.

And I think this really set the stage for subsequent studies after that. This really has established posaconazole as the drug of choice for prophylaxis. We can argue a little bit about that in the coming session. But let's just say that that’s the baseline data.

I also wanted to point out that this study by Gomes was done at a single center, MD Anderson. It’s a retrospective review of 152 patients with newly diagnosed AML. And they found an actually quite high, sort of outlying incidence of mold infection in their group. Interestingly, this group found that in patients who are prophylaxed with the echinocandins, there were more breakthrough infections with mold infections. They also did a lot of clofarabine treatment with reduced intensity transplants. They ended up having a lot of mold breakthroughs there, as well, so there were a number of confounders in their population, making definitive interpretation difficult.


So a more contemporary study comes from Italy looking at a registry study from 2010 to 2012, done prospectively in AML patients who are followed from pre-hospitalization to day +90. They looked at incidence of IFI, as well as risk factors and found that in 712 patients going through induction remission, about 60% of these patients had received posaconazole, 25% had a secondary leukemia, and these leukemias are notably difficult to put into remission. And their overall incidence of proven and probable IFI was 6.1% by day 30. So this is a little bit more recent data to look at in terms of what the baseline would be.

This is an interesting study because they went on to look at pre- and post-chemotherapy risk factors for IFI, especially mold infection, in particular.


But they, not surprisingly, found that COPD and ECOG performance status of greater than 2 were linked to proven and probable mold infections. But they also found that house renovation in the 6 months prior to treatment and a high exposure job, meaning things like farming or working in a floral shop or construction work were also associated with subsequent IFI due to molds.

This is the most puzzling piece, and that is that having a higher body weight was relatively protective. And by higher body weight, they meant a BMI of 30, which they really had trouble explaining. So before you reach for the glazed donuts, I'm not sure how to really interpret that data. But at least, these were not cachectic people certainly. Post transplant, not surprisingly, they also showed modest association with longer neutropenia durations, esophagitis, grade 2 or more as being predictive. And then posaconazole prophylaxis, as we've seen earlier, was relatively protective.


In the allogeneic transplant situation, the largest study to date has been from the TRANSNET Group, which was a consortium of 23 US transplant centers, who prospectively collected data between 2001 and 2006. Again, prior to the widespread use of triazoles – looking at 875 stem cell transplant recipients. They found overall a 3.4% incidence of IFI in that group within the first year after transplant. But that, as you'd expect, the incidence varied by transplant type. And I alluded to this earlier, but autotransplant shown in the dash line at the bottom, 1.2%.

So really this is not an issue, as you well know, of autotransplant patients. But it really varies by what type of allogeneic transplant is performed, with the unrelated donor and mismatched transplants occurring at rates of about 8% at 1 year, and 5.8% in those who are matched and related. Again, Aspergillus predominated, accounting for about 40% of IFIs; Candida representing about a third, not quite; and zygomycoses, 8%.


So again, in a more recent study from Italy, risk factors for IFI after allotransplant. This was a prospective study amongst 30 Italian centers. Almost 2,000 stem cell recipients, and about 14% received a mold-active agent for prophylaxis. They found several categories of IFI early in the course before day 40, with a cumulative incidence of 5.1%. And risks were unrelated or cord blood transplant or having a prior IFI or having acute leukemia at transplant. I think we could have predicted many of these.

And then late IFI, that is after day 40 to day 100 – just a small percentage, not quite 2%. Again related to having had an unrelated transplant or a cord blood transplant or acute GvHD with concomitant methylprednisolone or its equivalent. Regarding receipt of a mold-active agent for prophylaxis – and remember, this is in an allotransplant group, but those who received a mold-active agent had a reduced overall incidence from 9 to 4.6%. Again raising the question, is that important? In other words, should we be using mold-active agents in the post-transplant setting? Or is fluconazole enough? And I've listed several of the other risk factors that have emerged, as well, from other studies.


But I want to go to that question that I just asked. And that is, what about the use of mold-active agents in the setting of allogeneic stem cell transplant? And here is the classic data that was published by John Wingard in 2010, from a randomized double-blind trial of fluconazole—no activity against invasive molds like Fusarium, etc. versus voriconazole, which does have that anti-mold activity.

For prevention of IFI after allogeneic stem cell transplant, they provided either fluconazole or voriconazole for the first hundred days after transplant. And in some situations, those who were felt to be at higher risk were able to continue on their assigned azole therapy, which the hypothesis that if you give people voriconazole, something that’s a broader spectrum azole, you would decrease the incidence of IFIs. You would presumably decrease the incidence of Aspergillus infections, have less need for empiric antifungal therapy, and have greater fungal-free survival. I think that’s sort of an intuitive hypothesis.

The P values and the incidences are shown here. There was no decrease in IFIs. There was a trend toward fewer infections in those who only received fluconazole. There was no difference in terms of empiric antifungal therapy, and no difference in fungal-free survival. So I think at this stage, most of the transplant centers in the US and elsewhere use fluconazole primarily for prophylaxis in this setting.


I also want to show you another pivotal study for allotransplant patients with GvHD. This was the posaconazole versus fluconazole study, published by Ullmann in the New England Journal in 2007. And I show it even though it’s older data; it’s important because it serves as the basis for what we're going to talk about today. And in this study, patients who had a severe GvHD and were on at least 1 mg/kg of methylprednisolone or equivalent daily were randomized to receive posaconazole or fluconazole prophylaxis. And as you can see here, in the first 100 days, those with a greater probability of infection due to invasive fungal infections were in the fluconazole group, with proven or probable invasive aspergillosis being decreased significantly from 7% to 2.3%, although there was a trend toward decreased overall IFI. But most importantly, there were mortality benefits seen in this study – 4% versus 1%, with the benefit seen due to posaconazole.

So talking about mortality, if we look at some of the older data, this is where we really I think all of us become very nervous about invasive fungal infections. And that is the fact that they tend to be lethal in our patients.


These are data again pre-triazoles with having an attributable mortality of about 40%, down from the 1990s when it was more in the 60 to 70% range. And there are some data to suggest that it’s decreased even further in the current era, certainly. For Candida species, the mortality rate is about 33%; zygomycoses and Fusarium are the ones that we really have most concern about because of the associated very high levels of mortality. A number of factors are associated with mortality. And so, in our patients with prolonged neutropenia, some renal or hepatic insufficiency, methylprednisolone use and the idea that delayed diagnosis and delivery of antifungal therapy contribute to mortality, are all factors that promote the use of voriconazole and posaconazole, and perhaps isavuconazole – as we’ll see. These are the factors that really have all of us so concerned that we end up prescribing those agents frequently.

And I want to share with you this kind of apocalyptic quote from some folks who wrote in Clinical Infectious Diseases about a year ago, including John Wingard:


That’s what we're here today to talk about. How can we better use these drugs to improve the outcomes of IFI in our hematologic malignancy and allotransplant patients? Specifically, what new diagnostic methods will be available to us to more accurately and rapidly define IFIs? And how should we use the new triazole drugs so that we avoid spiraling empiricism? What are the advantages and problems associated with each of the ones that are available? And finally, is azole resistance really an emerging problem? Is it something that we have to be concerned about in our patient population?

And so with that, I'm going to turn it over to Dr. Maertens and Dr. Lewis to start answering some of these questions for you.

SECTION 2: IFI DIAGNOSTICS

Johan Maertens, MD, PhD

Although I'm a hematologist by training and a transplant physician, I've been asked to talk about the currently available and emerging tools that we have for diagnosing invasive fungal infections in hematology patients. And I will also address the question of how can we incorporate and how can we integrate these new diagnostic tools into our diagnostic and therapeutic algorithms.


We’ll talk about fungal diagnostics, what is currently available. I will not discuss radiology, although radiology has been a key element or has become a key element for diagnosing invasive mold infections. But I will only focus here on what's available from the mycology or the microbiology lab. And in the interest of time, we will focus on mold infections and not on yeast infections. So, first of all, we have a number of direct tests or conventional tests. And these are histopathology, direct microscopy, and culture. Now histopathology has always been one of the key elements and has always been the gold standard for making the diagnosis of invasive mold infections, also in hematology patients.

And as already mentioned by Alison in her talk, that has been acknowledged by the EORTC/MSG definitions, where you see that histopathology is really the tool that we use to make a diagnosis of proven fungal infection. But you also know that both in clinical practice and in clinical studies, we don’t have that many patients with proven disease, most of them have probable disease. And then we have culture and direct microscopy. Direct microscopy, often by the use of optical brighteners, such as Calcofluor.

Now there seems to be some problem with culture. Culture is taken from normal, sterile sites. First of all, culture has a low sensitivity. This is certainly true for blood cultures, if you talk about invasive mold infections. Usually our blood cultures are negative, although there are few notable exceptions, like Fusarium and others. But, if we talk about invasive aspergillosis, in 99% of your cases, blood cultures will certainly be negative. So there is low sensitivity in hematology population, sensitivity is at best around 30% to 50%. And also the predictive value of a positive culture depends on the underlying condition of the patient. Another problem with culture is that it is time-consuming. These fungal pathogens grow slowly in the different media that we use. And it takes time to identify these fungal pathogens.

However, there are also a number of advantages. If you have a positive culture, you can do susceptibility testing. And this is important in the context of growing resistance or emerging resistance, especially azole resistance. And, often, culture is the only way to detect multiple fungal infections in a single patient.


Now over the last couple of decades, I have to say, we have seen an increase in the number of nonculture-based diagnostic tools. Among these indirect tests, some focus on mold infections, others more on yeast infections.

And we have, for instance, galactomannan detection that has become available now. We can look for anti-mannan for detecting invasive Candida spp. infections. There is a T2 assay that is available in this country as well as 1-3 β-D-glucan, which is more of a panfungal assay. We’ll discuss more about that later. As already mentioned, PCR may be available – a panfungal PCR or a species-specific PCR. And then there are the new kids on the block, the lateral flow assays, both for detection of Aspergillus infection, but also other non-Aspergillus mold infections.

As you can see, the list is not complete. You do not see tests for cryptococcal infections on the slide. And also, important for this country, we will not discuss tests for the endemic mycoses. So again, we will focus here mainly on the mold infections.


We will discuss what can be routinely available in many centers, that is detection for galactomannan, detection for 1,3-β-D-glucan, and PCR.


Now let's start with galactomannan. If you look at the composition of a fungal cell, first of all you have the fungal cell membrane, which is composed of a bilayer of phospholipids. And on top of that, there is a fungal cell wall. And that fungal cell wall has different layers of molecules, be it mannoproteins, glucan, and chitin. Now these mannoproteins are really consistent of different manno residues held together, and they have different side chains. The test that we use to detect galactomannan is a commercially available test by Bio-Rad Laboratories. It’s called the Platelia Assay. And it uses a monoclonal antibody, that’s a rat monoclonal antibody, EB-A2, that binds to the immunodominant epitope in galactomannan, and that’s basically the side chain. Here you can see the galactofuranoside chain. Now this side chain is not specific for galactomannan. You will find it also in other different mannoproteins. So basically what you detect with these tests is not really galactomannan, but macromolecules containing galactofuranoside as a side chain.

We know that galactomannan is secreted only by growing hyphae. It’s not secreted by resting conidia, only by growing hyphae. And so, galactomannan is really released into the circulation when the fungus becomes invasive. The epitope can be found on many different body fluids. Usually we use blood samples, be it serum or plasma samples, for testing for galactomannan. But it can also be performed on urine. It can be performed on cerebrospinal fluids. It can also be performed on BAL fluids, abscesses, pleural fluid, etc. So basically any body fluid can be tested for the presence of galactomannan.


Now this is how the test works. It’s basically a sandwich ELISA. You have a microplate that is coated by the rat monoclonal antibody. And that is used as a capture antibody. If you add a clinical specimen, and if there is galactomannan present in that specimen, galactomannan will bind to the monoclonal antibody. And we use that same antibody as a detector antibody. So now the galactomannan is really sandwiched in between the two monoclonal antibodies. And then we add a chromogenic substrate, and then the test can be read by optical reader.

What the test gives you is a ratio. So it doesn’t give you the actual amount of galactomannan, but it gives you a ratio. And this is calculated by dividing the optical density of the patient sample by the mean optical density of a control sample that is provided by the manufacturer. And so, it gives you an optical density index.


What do we know about the specificity of the galactomannan assay? Well, it’s rather specific for Aspergillus spp. But there is some cross-reactivity. Patients having invasive Fusarium spp. infections, for instance, may be galactomannan positive. The same is true for Acremonium and for Histoplasma spp. We don’t have Histoplasma in Europe, but I know that you see it in this country.

So you have to take that into account, that patients with Histoplasma infection can be GM positive. And also the patient infected with Alternaria and Penicillium spp. can be GM positive, although Penicillium is usually considered a contaminant coming from the lab, and not really a human pathogen. And there are fewer exceptions listed over here.

Usually GM is negative when you have a patient with a Mucorales infection. I say usually because we've seen a number of patients that were, indeed, galactomannan-positive. And the difficulty here is to see or to assess whether this is cross-reactivity between Mucorales, or whether these patients really have dual infection – Mucorales with Aspergillus. The tests will always be negative in case of invasive Candida spp. infections, invasive cryptococcal infections, or pneumocystis infections.


Now before you start using this test or before you start ordering the test, you have to know that the performance of the assay is influenced by a number of biological and epidemiological factors. And without going into too much detail, not looking at all the elements here listed, we list some of the biological factors that clearly play a role in the performance of the GM test is the site of infection.

If you have an encapsulated infection, like an abscess, then there may be no release of galactomannan in the circulation. So you may have what is called a false-negative assay, even with a patient with documented infection. Clearly there is an impact of exposure to antifungal agents. And I have to say, an exposure to mold-active antifungal agents. Many of them are mold-active azoles, but also the mold-active polyenes – and even the echinocandins. If there is exposure, be it prophylactically or empirically, that will decrease the sensitivity of the assay.

And you also have to look at the underlying condition. The performance of this test is best in severely neutropenic patients. It performs less well in the non-neutropenic hematology patients. And it performs poorly in the non-hematology population. So if you use this test in solid organ transplant recipients, in HIV patients, in ICU patients, sensitivity will be poor. However, if you focus on the prolonged neutropenic hematology patients, then the sensitivity is around 80 to 90%. And there are a number of epidemiological factors that play a role, such as your sampling strategy. Do you ask for the test only once a week? Or do you ask for it twice or three times a week? That will have a major impact. Your underlying prevalence of invasive fungal disease, and of course, the cutoff that you use for positivity will have an impact on sensitivity and specificity of the test.


Looking at β-D-glucan, as already mentioned by Alison, 1,3-β-D-glucan has been included in the revised EORTC-MSG Consensus definitions, as well as the microbiological criteria for diagnosing probable invasive fungal disease. And it is a major cellular component of most fungal species, not of all fungal species – and I’ll come back to the specificity in a minute.

There are a number of different commercially available assays that can be used. We normally detect 1,3-β-D-glucan using blood samples. It can also be performed on cerebrospinal fluids. But you have to remember that, for instance, a BAL sample is not suitable for this assay.


Now different tests are commercially available. However, if you look in Europe and in the US, only the Fungitell test, which was formally called the Glucatell test, and is manufactured by Associates of Cape Cod, is now approved by the FDA and is also available in Europe.

All the tests, such as the Fungitell-G, the Wako test, and the Maruha test are mainly – not to say exclusively— used in Japan. They are not available in this country, nor in Europe. Importantly, if you read the literature on this test, you have to know that a cutoff that is used to define a positive assay is different for all these different assays. And it’s around 60 to 80 pg/mL for the Fungitell assay, and significantly lower for the Japanese assays.


Now this is looking at the specificity with 1,3-β-D-glucan assay. You will detect Aspergillus cases, Candida cases, but a lot of other fungal pathogens will be detected, as well, including Fusarium, Trichosporon, Acremonium spp., and all these listed here on the slide. Also, Pneumocystis jiroveci will be picked up by this panfungal assay. And it’s actually a very nice test to make a diagnosis or to improve the diagnosis for Pneumocystis jiroveci pneumonia.

The test is usually negative for cryptococcal infections. In the early days, we said that infections with Cryptococcus spp. always yielded negative 1,3-β-D-glucan results. Now we've seen a number of cases that tested positive. So that’s why I said it’s usually negative. And you will not pick up infections caused by Zygomycetes with this test.


Now, for both the β-D-glucan assay and the galactomannan assay, there are a number of false-positive assays – false-positive, but also false-negative assays. I've listed the reasons here.

The use of some β-lactam antibiotics, such as piperacillin/tazobactam or amoxicillin/clavulanic acid, can result in false-positive galactomannan assays, as well as false-positive β-D-glucan assays. So, you have to remember that if you use BAL fluid, and if it is very sticky, and you add a mucolytic agent before you use it in the lab, some of these mucolytic agents can result in false-negative galactomannan assays.


Now only one word about PCR. You have seen on the slide presented by Alison regarding the EORTC-MSG definitions, that PCR is still not added to one of the microbiological criteria. And that’s because PCR is really not standardized. Many of us use PCR. There are a number of commercially available PCRs for detecting Candida, for detecting Aspergillus spp. They are not really popular, at least not in Europe. And many of us use in-house developed PCRs, but as already mentioned, they are not standardized. And as such, they have not been included as microbiological criteria.

However, there is a great initiative, at least in Europe. But there is collaboration also with centers in the United States and in Australia. It’s called the Europe Aspergillus PCR Initiative. That was launched in 2006 under the umbrella of the International Society of Human and Animal Mycology. And the aim of that initiative is really trying to standardize the whole procedure of PCR.


And then finally, there is the lateral flow device, which is really a point-of-care test if you use BAL samples. It’s not a point-of-care test if you use serum samples or plasma samples because there is a pre-heating test that needs to be performed in the lab. But if you use BAL samples, it really is a point-of-care test that can be done at the bedside. And it takes about 15 minutes to have a positive or a negative answer.

This test also uses a monoclonal antibody. In this case a murine monoclonal antibody, different from the rat monoclonal antibody that is used in the galactomannan assay, recognizing a constitutive glycoprotein antigen. That antigen is secreted during active growth of the hyphae, so it’s not secreted by resting conidia. And as you can see, it displays superior specificity to the rat monoclonal antibody that is used in the galactomannan assay. And this test has been developed as a rapid, and certainly user-friendly, diagnostic test.

Here you see some aspects of the specificity. So this is what the assay looks like. And here we've tested 5 different fungal pathogens. This is the assay that was specific for Aspergillus fumigatus. And if the test is positive, then you see a second band appearing in the assay.

Now the question is how can all these diagnostic tools help us in our clinical practice?


These assays can help us in different ways. They can help us to exclude the diagnosis of invasive fungal disease. And if you want to use the tests for excluding the diagnosis of invasive fungal disease, you certainly rely on the negative predictive value of these assays. And you use the negative predictive value either to stop the antifungal treatment that was started empirically or prophylactically, or to withhold antifungal therapy. Most of the time we use the assays in this way in our clinical practice.

The assays can also help us to support the diagnosis of invasive fungal disease and this is the way we use the assay mostly in clinical studies. To support the diagnosis, we rely on the positive predictive value, either to ask for additional diagnostics—for instance, to ask for a CT scan, or to start early antifungal therapy. And these assays can help us to predict outcome. And in that case, we rely on the baseline values or we look at the kinetics of the assay to choose or to modify our antifungal therapy.

Now there are a number of conditions that we have to take into account. And the first one and probably the most important one is that for a given sensitivity and specificity of the assay, the test performance is mainly driven by the pre-test prevalence of the target condition. And this is a common theme, and we’ll come back later when I talk about prophylaxis.

We should also look at the importance of antifungal prophylaxis—again, something I will discuss within a minute. There is the importance of the strategy of using these assays in a surveillance strategy or for early diagnosis. The importance of the sample that we are going to use, a serum sample or plasma sample, or a BAL fluid. And the number of samples that we use will have importance, as well.


So if you use these biomarkers as screening tools—again we rely on the negative predictive value. So you are looking for tests with a high sensitivity. You don’t want to have false negative assays. Now this is looking at a number of different meta-analyses that have evaluated performance of the galactomannan assay, the β-D-glucan assay, and PCR. And you see that for the galactomannan, the sensitivity is around 80% in meta-analysis. For PCR, it’s around 85%. It’s important to note that we have a high sensitivity, despite the fact that this is not the standardized assay. And if you look at β-D-glucan, it’s somewhat low. It’s around 70%.


However, if we start combining these assays, then clearly you have different results. This is again a meta-analysis looking at GM and PCR as used as screening tools in high-risk hematology patients. PCR alone has a negative predictive value of around 96%. For GM, it’s 98%. If we start combining GM and PCR as screening tools, the negative predictive value is 95%. But as you can see, you clearly increase the positive predictive value up to 88%.

Now these are data taken from meta-analyses. If you look at probably the most important study that has been done, it was published by Rosemary Barnes in the Journal of Infection a couple of years ago. In that study, they looked at high-risk hematology patients. These patients were screened weekly for galactomannan and PCR, twice weekly. And the results of the study are that if you look at these tests individually, the sensitivity and the negative predictive value are OK. But if you start combining these tests, when you take the results together, GM and PCR, then you have a negative predictive value of almost 100% with a very good negative likelihood ratio. Clearly, saying that you can rely on the negative predictive value of this combination to withhold empirical antifungal therapy.


Now we can also use these biomarkers as a diagnostic tool. And if you use them as diagnostic tools, they are mostly performed on BAL fluid. And in that case, you want to have a high positive predictive value. So you're looking for a test with a high specificity. You want to be sure that a positive test result really means that you have invasive aspergillosis.


Again looking at different meta-analyses here – and here, the question is what is the cutoff that you are going to use? Is it 0.5? Is it 1.0 or 1.5? And if you look at the meta-analysis, 1.5 seems to be the most appropriate cutoff, resulting in a high diagnostic odds ratio of 674. So the conclusion of this meta-analysis was that a BAL galactomannan optical density index of 1.5 or higher is highly suggestive of invasive aspergillosis, whereas a negative result virtually always excludes invasive aspergillosis.

So if you use an assay, you have to know that the same assay – that maybe you have to use different thresholds. Clearly if you use galactomannan for screening purposes, your threshold will be 0.5. If you use it for diagnostic purposes, your threshold will be 1.5.


One final word about antifungal prophylaxis. Kieren Marr clearly showed in a study published in 2005, that if you use mold-active agents— azoles, polyenes, and even echinocandins—you decrease the sensitivity of the galactomannan assay. And the same is true for β-D-glucan and probably also true for PCR. And why is that? Well first of all, because mold-active agents decrease the baseline incidence, and so they will also decrease your positive predictive value.

Now one of the questions is can you still rely on serum galactomannan if you give mold-active agents to your patients? And we just heard from Alison that many of us use posaconazole prophylaxis now. So can you still rely on serum galactomannan in posaconazole prophylaxed patients?

Well, yes you can. But then you have to use it not as a screening tool, but as a diagnostic tool when you have a high suspicion of invasive fungal disease. And that’s very nicely shown by a recent study published by Rafael Duarte, a study from Spain looking at the performance of serum galactomannan in high-risk patients receiving mainly posaconazole or voriconazole.

If you use it as a screening tool in these patients, as already mentioned, the incidence of invasive aspergillosis is going to be 2% or lower. You still have a high negative predictive value, but the positive predictive value is only 12%. However, if you have a high suspicion of invasive fungal infection—so a breakthrough fungal infection in a patient on mold-active azole, then you use it as a diagnostic tool—your baseline incidence then is going to be much higher; it’s around 50%. And then your positive predictive value rises from 12% to 90%. So it can still be used—not as a screening assay, but as a diagnostic tool.


So finally we have to try to incorporate these new diagnostic tools or these biomarkers into our antifungal management strategies. We know that we can give an antifungal prophylactically or empirically. Empirically really means that you have a fever-driven approach and that you give it to your patients with unresponsive fever.

Also we have directed treatment if you have proven probability cases. But now with the new treatment approaches, we start relying on these new diagnostic tools, we incorporate them and we may have more diagnostic-driven approaches where we decide to start an antifungal agent on the basis of a positive biomarker, or on the basis of a positive biomarker in combination with non-typical pulmonary infiltrates seen on a chest CT scan or on a standard radiology.

SECTION 3: IFI THERAPEUTICS

James S. Lewis, II, PharmD

One of the things that has gotten really interesting is that any time I wade into the oncology area—because I'm kind of an ID person normally—I get serious drug envy. And as a pharmacist, you can kind of see where this comes from. I get drug envy very easily. I want to tell you guys—I got off the plane last night, went down to baggage claim. I walk out to baggage claim, there is a sign there for a TKI. I drive down the highway, there are two billboards on the way here for a TKI. You guys are getting new drugs weekly.

We in the ID world, we're lucky if we get a new drug once every year or two. And so, we're still going to be talking about data with drugs that’s a few years old here, and that’s probably mind blowing for you guys in the oncology world. We're still talking about voriconazole from 2002.


This is the FDA indication list for the antifungal agents that we currently have available on the market. And I note the key phrase here: FDA approved. This is not how they're necessarily used clinically. But this is what they are approved for by the agency. And what you notice really quickly here is that there are some marked differences with regards to what these agents are actually approved for. We know at this point, as it’s already been shown a little bit, voriconazole has prophylaxis data. But it’s in no way, shape, or form indicated for that by the FDA.

The other thing to know about voriconazole now—and one of the reasons that this is going to be important is— insurance companies. Voriconazole is now generic in the United States. And what that has done to your price is roughly drive it down by about 75%. So now voriconazole is way cheaper than the other azoles that you see up here on this slide. And you'll see why that’s important as we move forward a little bit.

Posaconazole has only got prophylaxis data. You'll notice you've not seen treatment data up on this slide really anywhere. There's a little bit of retrospective treatment data. But again, this is with the oral suspension. The FDA in the package insert says that the tablets are the preferred formulation. When does the agency do that? Never, okay. So one of the things that you need to realize is that a lot of the data that you're seeing up there with posaconazole is with an oral suspension formulation, that it had to be the third Tuesday of the month after 4 pm to get reliable absorption. That’s how dodgy this stuff was. So we have really good data, even with an inferior product. And we're going to come back again to the different formulations with posaconazole.

That brings us to the new kid on the block – isavuconazole. But with isavuconazole, what you'll notice really quickly is there's no prophylaxis data yet. But the spectrums of these agents are so darned similar, guys, let me tell you, I really think that pretty much these drugs can be used for one another in different places.

And so, what you're going to see, I think going forward, is going to be largely driven by financial implications: voriconazole being generic, being a big one. These two (isavuconazole and posaconazole) are going to be branded for a while. And so, I think you're going to see a bit of a price dogfight potentially break out between the two of them.


Alright, so what about amphotericin B? We're talking a whole lot about azoles up here and on the previous slide. Why are we spending so much time on azoles? Well again, coming back to ancient data circa 2002, I just want to remind you really quick that voriconazole took amphotericin B out and kicked it around the block for invasive aspergillosis.

So basically we woke up on Thursday morning before the New England Journal of Medicine was published. And if I walked up behind you in the hall of your medical center and said invasive aspergillosis, you would have immediately said AmB. That’s what you would have done. That night, we went to bed after the publication of this data and the answer switched immediately to voriconazole. The interesting thing was how fast the change occurred. It says how much all of you in this room hated AmB. Because I have never seen uptake of clinical data so quickly in my life. Our hematologist ran screaming from the room with regards to AmB. And there was a party now that they had something else to use for invasive aspergillosis. So we don’t talk a whole lot about AmB because voriconazole flat out whooped it, to use a term that we would use in Texas a lot.


But what about liposomal amphotericin B? It’s still out there. It’s still the broadest spectrum game in town. And we know it’s better tolerated than the old school, which should be put in a box and dropped in the middle of the ocean. So are we still using it? Yes, we are—especially in people who can't tolerate the newer generation azoles, or God forbid, have breakthrough infections while on one of these newer azoles. And we're not supposed to do this, but we all do it. When you look at the comparison of response numbers across the different groups here, these numbers, as far as response rates go for liposomal amphotericin B, look very similar to voriconazole.

We've not seen head-to-head data between liposomal AmB and any of the current generation of azoles out there. But these response rates pretty much look in line. So we do still use some liposomal AmB, but more so in an intolerance or salvage role than probably anything else.


What about echinocandins? We all absolutely love echinocandins. And the reason for that is that they're so darn well tolerated. From a pharmacist's standpoint, the lack of drug interactions, these drugs are almost boring sometimes. I mean, they don’t quite give me enough to do as a pharmacist. And so, the echinocandins are really awesome, but the downside of them is the spectrum is atrocious. They get Candida and they get Aspergillus spp., and that’s about it.

So they're being used a lot more by you guys for prophylaxis in the intense AML induction chemotherapy setting, in order to get away from some of the drug interactions with these azoles. And guys, that’s really, as you're well aware, the major problems with the azoles—we're seeing a lot more echinocandin use pop up.

The slide shows data out of MD Anderson that looked at people who are receiving induction chemotherapy. And what they saw was they got a lot more breakthrough fungal infections when they got an echinocandin than they did with one of the newer generation azoles. Now when you really drill down on this data, one of the things that's very interesting is that a lot of the breakthrough data, especially on the echinocandin side, was yeast based, which really surprised me. I expected it to be more driven by mold breakthrough. But this difference is largely driven by a lot of yeast breakthrough. So I think that’s something to kind of think about. And again, as was mentioned previously, this is single center data. So we need to see a little bit more of this. But this really kind of gives me a little bit of reason for concern. And I’ll tell you, our group in Oregon, we really try to keep the new generation azoles out there as much as we can in this situation.


So when you're talking about what you want to see from a perfect antifungal agent, you've got a long shopping list here. And the problem is that when you go through the shopping list you realize very quickly, we don’t have the perfect antifungal available to us.

Each of the three classes, the polyenes, the echinocandins, and the azoles, all have significant challenges when you look at this wish list on this slide. And so, really we're in an area where we're having to kind of give-and-take a little bit with regards to the strengths and weaknesses of each of the compounds we have available to us.


So with that being said, let's talk a little bit about the azoles. And we're going to spend the bulk of our time for the remainder here talking about azole therapy. Now LFTs drive us all nuts. I think everybody in here has seen the person who you started on whatever “aconazole,” and then a couple of days later their LFTs start to move. But the problem is that you guys specialize in using drugs that cause LFT abnormalities. And then you do things to these patients that cause this lovely disease state called graft-versus-host disease, which I've seen also do some very fun things to LFTs. So it’s really kind of a multifactorial problem. But we know that within the class, all of these azoles do it. And it’s not necessarily dose related. And that’s really beautifully and best laid out in the fluconazole package insert.

The million-dollar question is here in bullet 3. What's the cross reactivity? We don’t know. And so I think it is certainly worthwhile if someone’s LFTs are moving—especially this means you, voriconazole—to try a different azole and see if you can back down the LFTs a little bit. And when and where do you discontinue this? Again, it’s kind of a pain threshold issue because there's really no clear guidance out there as to at what elevation of LFTs should we be thinking about moving to the potentially nephrotoxic LAmB, or to the much more narrow-spectrum echinocandins? So it really does become a very, very challenging dance to do. And we really think voriconazole is probably the worst of the bunch with regards to LFT abnormalities. But again, the new formulation of posaconazole—I would argue that a lot of the reason that we never saw LFT abnormalities with the oral suspension was because of variable absorption. You're not going to see LFT abnormalities with an undetectable posaconazole level when there's no drug there. So I think we're about to learn a lot more with the new tablet and IV formulations of posaconazole about what the true incidence of LFT abnormalities with posaconazole really is. And we're really going to get a good snapshot here really quick with the isavuconazole versus voriconazole data to look at how those two stack up head-to-head.


This is a big moment in my life. I grew up as a Star Wars geek. So imagine my pure excitement when several years ago I walked into ICAAC and saw two of my favorite things in the same place—fungal infections and Star Wars. Sitting in the middle of the poster at ICAAC is Chewbacca. And the reason that Chewbacca was sitting in the middle of the poster: this was a case series out of the NIH, talking about hallucinations due to voriconazole.

The paper, which was later published, and the poster that really did a nice job of laying this out, talked about a gentleman at the NIH medical branch who was receiving voriconazole. The medical team walks in and he’s like— guys, there's something I need to tell you. At least this is how I pictured this conversation going. There's something I need to tell you. Chewbacca is on the ceiling. I know Chewbacca is not on the ceiling; he’s not really there. But Chewbacca is on the ceiling. And this is really one of the issues with voriconazole. We've learned over time at these higher serum concentrations, especially over about 5.5 mcg/mL, patients start to hallucinate.

We had a guy at UT San Antonio thank us for the lovely artwork at the foot of his bed and tell us how the artwork changed every day and wanted to know if he was going to be charged for that, by the way. But there was no artwork at the foot of his bed. His voriconazole level was 9 mcg/mL. This is the kind of stuff that we have seen.

We've also seen an increasing discussion of skin cancer, fluoride problems, LFT abnormalities, and—the always awesome—erratic pharmacokinetics that you see with voriconazole. So let's talk a little bit more about the voriconazole tolerability issue.


This skin cancer issue is bothering me more and more the more I look at this. And really, it appears to be dose and duration dependent. And what they think – although they're not totally sure – is that it may be driven by the voriconazole main metabolite, which is voriconazole N-oxide. And the issue I think is that a lot of this data has been in the dermatologic literature. Some of it has been in the oncology literature. But it’s not all been focused in an area where a lot of ID and oncology people are necessarily seeing it all the time.

The other thing that we have had to deal with a lot more is this fluoride issue. We learned a lot with the Exserohilum outbreak after the contaminated steroids out of Boston, where they had shot steroids full of fungus into people’s central nervous system. We learned a lot about high-dose voriconazole over the long term. And one of the things that we saw is with this trifluorinated structure, that you were getting fluoride serum levels in patients that were considerably higher than you would have liked. This was associated with a lot of bone pain and a lot of periostitis, especially in the ulna and in the ribs.

So we've got skin cancer issues. We've now got some fluoride issues. And we certainly have the hallucination stuff that we talked about earlier. One of the things also that came out was that there's also a lot of alopecia that occurred in the patients who were getting higher doses of voriconazole during the Exserohilum outbreak. I want you to stop for a minute and think about Chewbacca with a little alopecia.


Then finally, as if this isn't getting complicated enough, Pascual and colleagues come out a couple of years ago and said everything that you know about the bioavailability of voriconazole is wrong.

When voriconazole came out in 2002, one of the strengths of the compound was that it was supposed to have 100% oral bioavailability. But I think as increasingly a lot of us have used this drug, we've realized that it’s not one-to-one. And Pascual was really the group that came out and clearly showed us that the bioavailability, especially in sick oncologic patients, is probably in the 60 to 80% range. And so that bioavailability difference makes the dosing again a little bit dicey. And we haven’t even gotten in to all of the P-450 issues with regards to voriconazole yet. Look at this study if you've not looked at this before because, the bioavailability is not as good as many people think. And that bioavailability issue, especially when you're doing therapeutic drug monitoring, really comes back to get you.


Now posaconazole, I want to say a couple of things about posaconazole. First – thank you. The IV and the tablets are awesome. It is so nice to have a drug that finally gets in, that you don’t have to mix it with olive oil, orange juice, and stand the patient on their head in order to get them to absorb it.

The new tablets are flat out awesome. Albeit a little bit big for some patients. And the IV is also awesome, but it did come with cyclodextrin – but beggars can't be choosers, right? Now the major problem with posaconazole at this point in time is that it’s costly compared to voriconazole (which has become generic): $155 a day for tablets, or $500 a day for IV. By comparison, you start talking about voriconazole being at about $40 a day, the IV is up around $100 a day. You're talking about considerably more expensive when it comes to the posaconazole formulations that are out there.

Also, there have been an increasing number of reports of dose confusion. Realize with posaconazole that the tablets and the IV are dosed 300 mg once a day. Not the 200 mg 3 to 4 times a day that you guys are used to with the old oral suspension. As a matter of fact, the package insert for posaconazole was re-updated to kind of remind you of that fact. And there have been a couple of case reports. One in a kid where the oral suspension dosing was given using the tablets and you got crazy high levels and some toxicity. So I think this dose confusion thing with posaconazole is really something you want to be aware of and remember that it’s once a day now with the tablets and the IV formulation.


The issue with isavuconazole at this point in time is that we don’t know what we don’t know. If you go backwards about 15 years ago and look at the development of posaconazole and voriconazole, what we were told at that point in time was, “Nope, no, nope. No need for therapeutic drug monitoring.” We've all heard this story right around 2000. We're hearing the same story on isavuconazole. But we've not really had a really great look at all of the pharmacokinetic data yet. And so, I'm going to withhold judgment on what type of therapeutic drug monitoring is really going to be necessary here. Because again, this is going through P-450 systems just like posaconazole and voriconazole. There's variability in those P-450 systems just like posaconazole and voriconazole.

It does appear that it’s possibly a little bit better tolerated than voriconazole. And again, it’s once daily after two days of loading. The nice thing about isavuconazole is it’s got a crazy long half-life of about 380 hours, I believe. And so it allows you to really get on that once-daily dosing. But that load goes over two days.

It’s again IV and oral. And again, it’s 200 mg both IV and oral with the 1:1 bioavailability. The interesting thing here though is to be aware, this comes as a pro drug. The pro drug on this is isavuconazonium sulfate. There's also again no cyclodextrin in the IV, which is a nice advantage because the pro drug is very water soluble. And it only has the treatment indication at this point. Remember it’s indicated for the treatment of invasive aspergillosis, as well as the treatment of invasive mucormycosis, albeit with only about 37 patients in that Mucor treatment data set.


So here again, coming back to one of the questions that we had earlier is the voriconazole versus isavuconazole phase 3 data in invasive aspergillosis. What you see is really no difference around a lot of the major endpoints down here. But where you do see it is the drug-related adverse events.


What's really different as far as the adverse drug events go is really areas that you would kind of expect. We know historically that voriconazole has had issues with skin. Again we're talking about the skin cancer thing here.

Again coming back to voriconazole really quick – the FDA updated the package insert in February of this year. Again kind of beefing up the wordage on skin abnormalities, visual abnormalities, as well as some of the LFT stuff. So really I think the challenge with voriconazole continues to be the tolerability of the compound. And really the issues within the variability of the PK.

So when you look at isavuconazole head-to-head with voriconazole, you would hope to see these kind of three common problem areas look a little bit better for isavuconazole. And at this point, they really kind of do. It’s important to note though, and thank you to Johan for pointing this out to me prior to today, was that the hallucinations are not included in the visual disorders at all. That’s a separate breakout. And there really didn't appear to be a huge difference there, but that may be due to how the voriconazole was dosed, that you weren't really seeing super-high voriconazole levels there.


Now job security for me is on this slide. The drug interaction stuff with the azoles is one of the major limitations to the use of this compound. And really what you've got here are some of the major enzyme systems that are out there from a P-450 standpoint. Notice 3A4. Okay, 3A4 and 3A5 are really where the vast majority of drugs go through, including a lot of your oncologic drugs. And notice that all of these—look at all of them—are inhibitors. Now some of them are more potent inhibitors than others, with itraconazole and voriconazole being extremely potent inhibitors. With voriconazole, interactions are more likely with drugs metabolized via 2C9 and 2C19, but also with 3A4 inhibitors.

Posaconazole is a pretty good inhibitor, as is isavuconazole. So all of these are going to shut down your P-450 system and are going to push up the levels of anything that is being metabolized through those P-450 systems. So I think that Brüggemann, who's done a ton of work with this, really says it very nicely in a review that they published in CID going on about six years ago.

You've really got to stay on top of this, especially in your world. Because you're getting so many new drugs so fast that a lot of times the drug interaction really is not that well understood when a lot of these new drugs are coming to market. And when you start shutting down the metabolism of some of these fun-filled compounds that you guys are using, you can see some very substantial toxicities associated with them. And again, really trying to remember which drugs are living in which group. And I want to point out, notice that voriconazole is all over the place. Really voriconazole does multiple enzyme systems, and is all kinds of fun. But for P-glycoprotein, which I think is really underappreciated—posaconazole and isavuconazole are sitting out there. So, really, irrespective of where you go within the azole class, the drug interactions are a major challenge.


Regarding the tyrosine kinase inhibitors – again I am blown away by the number of these new molecules that are on the market. And what's really interesting – if you haven’t seen it, there's a great review on drug interactions with tyrosine kinase inhibitors. It was published in Lancet Oncology last year. And it’s really a good one to keep stuck in a filing cabinet somewhere. But when you look at these, a ton of these things— almost all of them—are going through 3A4. And when you're going through 3A4, you are going to bump into those azoles. You are going to do it. And so really, I think this is an area that you've got to pay huge attention to.

The other thing that’s very, very clear – when you look at a lot of the package inserts, the drug interaction work was done mostly against ketoconazole. Who cares? Nobody uses ketoconazole, and it behaves differently than the other azoles that we're playing with now. But it’s unfortunately kind of a gold standard in drug development. So really watch this.

I also want to shout out – always look at the package inserts, always look at your LexiComps, your Micromedex’s. Put your pharmacist in a headlock and drag them to go to drug interaction review with you. But also there's a very nice new website. It’s fungalpharmacology.org. There's a nice explanation of it in Journal of Antimicrobial Chemotherapy this month. And I played around with that website quite a bit. It’s really slick. But I think really having to stay up on the drug interactions with these new compounds is going to be a challenge for all of us.


Therapeutic drug monitoring – to wrap up really quick. If your patient is on voriconazole, you need therapeutic drug monitoring – period, end of discussion. And the reason for that is the variability of the compound. And also the fact that we know that below a serum level of about 1 to 1.5 mcg/mL, the drug doesn’t work. And above a serum level of about 5 mcg/mL to 5.5 mcg/mL, everybody sees Chewbacca. So you have efficacy issues and you have tolerability issues. And you have a narrow therapeutic index. Guys, that is the definition of a drug that needs therapeutic drug monitoring.

What about posaconazole? Posaconazole, it depends. If you're using it for prophylaxis and you're using the new tablet formulations, I think you can make a very strong argument for no therapeutic drug monitoring. We just finished an article on this– and it’s actually in press at Mycoses right now. Several other groups have done similar work. When you're using the tablet formulation, the incidence of patients having a posaconazole serum level that’s less than 0.7 mcg/mL is very low. Now if you're really worried about somebody’s GI integrity – they're throwing everything up, they’ve got rip-roaring mucositis, it might still be worth a look. But regarding routine TDM for posaconazole in the era of the tablet formulation, I'm not sure is necessary anymore. When it comes to treatment, I think I would definitely want them up around 1.0 mcg/mL, okay. But again, the volume of distribution for posaconazole is so high, it gets out in the tissue so well. And I think we're a little bit less clear because really posaconazole is just not been used for treatment that much. We don’t know a lot about it from a treatment standpoint.

And isavuconazole, who knows? You've got a massive volume of distribution. There's no signal in the phase 3 data. But then again, there's never a signal in the phase 3 data. It’s always as this stuff percolates out more. So we don’t really know with isavuconazole yet what the TDM requirement is going to be. We need to see a lot more PK data on isavuconazole to really help us make that decision.

In conclusion, the issue with the azoles is the drug interactions and the therapeutic drug monitoring considerations remain there. With isavuconazole, be aware that we don’t know what we don’t know about the pharmacology.

SECTION 4: PROPHYLACTIC, PRE-EMPTIVE, EMPIRICAL, AND DIRECTED ANTIFUNGAL THERAPY... STRATEGIES THAT ARE MUTUALLY EXCLUSIVE, OR...?

Eric J. Bow, MD, MSC, D. Bacteriol, FRCPC

Thank you very much. I would like to present an approach consistent with that one might take as an oncologist or as a transplanter, looking at the patient’s journey through the course of their illnesses. So, with that in mind, let me begin with a review of recent changes in medical mycology that may be relevant to clinicians.

First, the introduction and refinement of consensus-based definitions for invasive fungal infection gives us a common language around the robustness of the diagnosis. The term “possible invasive fungal infection” implies something different than “proven infection,” with implications for prognosis. These definitions were designed specifically for clinical trial use. Notwithstanding, clinicians have been applying them at the bedside.

Second, the availability of a wider spectrum of imaging techniques (high-resolution CT, magnetic resonance imaging, and PET scanning) has added to our abilities to use radiological features to define clinical syndromes involving body sites. Recognition of these syndromes is important because it helps direct microbiological and non-microbiological test strategies toward a more robust mycological diagnosis and therapeutic intervention.

Third, the diagnostic process has been enhanced by the availability of nonculture-based biological markers about which Johan just spoke. There is the galactomannan assay for species; however, a limitation is the production of this antigen by other molds in varying amounts. The β-D-glucan test for both molds and yeast, including Pneumocystis jirovecii, is nonspecific. The genomic techniques for mold and yeast based upon PCR may be more sensitive but remain nonstandardized. The use of mannan/anti-mannan for Candida spp. has limited usefulness.

Fourth, there are a limited number of classes of available antifungal agents for invasive infection. Each class has a different molecular target. Accordingly, there has been a renewed interest in the use of combinations, analogous to the principles employed for oncology regimens. Fifth, we have learned about the practical importance of drug-drug interactions, particularly with the azoles. The importance of the role of therapeutic drug monitoring in maintaining a balance between efficacy and safety has been discussed. Sixth, the recognition of risk factors for invasive fungal infection has served to target populations of patients for prevention strategies to mitigate those risks. The relevant risks are not continuous over a patient’s journey. The spectrum of patients at risk for invasive fungal infection has been expanded. We are transplanting a broader spectra of patients than in the past. The pre-test probabilities of invasive fungal infection, as Johan has indicated, have increased.

Four broad strategies of management have been developed; preventative strategies such as chemoprophylaxis; early therapy based on the detection of biological markers of infection, the pre-emptive approach; the empirical approach for clinical syndromes suspected but not proven to be of fungal origin; and directed therapies for fungal syndromes based upon the hierarchical classification of possible, probable, or proven infection. These approaches are not mutually exclusive. There is a great deal of overlap in how the available therapeutics are deployed. It begs the question about the value encompassed in these strategies for the resources that must be expended in their deployment.


This is a conceptual paradigm to help understand the relationships among burden of invasive fungal infection, the probabilities of infection control, and time. Oncologists are very mindful of the inverse impact of tumor burden upon prognosis. The relationship between fungal burden and outcome is similar.

The detection of surrogate biological markers based upon PCR or antigen detection techniques, together with high-resolution CT, may enhance the likelihood of early detection. Traditional bedside diagnostic tools based on signs and symptoms and conventional X-rays arguably detect invasive infection in patients late in the natural history of the disease, consistent with all-cause mortality rates in the range of 6-8 out of every 10 patients. The paradigm of early pathogen marker detection with therapeutic intervention has been employed for cytomegalovirus infection in solid organ and stem-cell transplant recipients as a standard of practice. The administration of antiviral therapy at the time of detection of CMV DNAemia prevents progression to clinical disease and the associated excess morbidity and mortality. This principle, based upon biological marker detection, is now being explored for invasive fungal infection in high-risk patients.


Chemoprophylaxis strategies are deployed among patients at high risk for invasive fungal infection but in whom there is no mycological evidence of fungal infection or clinical evidence of fungal disease.

A number of published studies in high-risk stem-cell transplant recipients, summarized in this mixed treatment comparisons meta-analysis, have demonstrated similar protective effects of the azoles against invasive candidiasis, but differential protection against invasive mold infections such as invasive aspergillosis. Second generation mold-active agents such as voriconazole and posaconazole appear to be more efficacious in the prevention of proven or probable invasive aspergillosis and for reducing all-cause mortality compared to either fluconazole or itraconazole. These treatment effects are encouraging; however, they remain imperfect since breakthrough fungal infections still occur despite these strategies.


The results of systematic reviews notwithstanding, it is encouraging that single center experiences continue to demonstrate similar treatment effects. Such is the case in this example in AML patients from Melbourne, Australia, where the mold-active second-generation azoles appear to have not only similar potent protective effects against proven and probable invasive fungal infection, but also a differential impact compared with fluconazole and itraconazole on the use of empirical antifungal therapy for the persistent neutropenic fever syndrome.


Adult patients with acute lymphoblastic leukemia who are receiving dose-intense multi-agent multi-cycle chemotherapy is a population at risk for invasive fungal infection that is often overlooked in clinical trials.

In this example from 4 hospitals in Melbourne, Australia, the anti-leukemic regimen–related event rates for proven/probable/possible invasive fungal infection seem similar to those reported for AML patients. These anti-leukemic regimens, like the childhood ALL regimens, are corticosteroid-intensive, are administered over many months, and are designed to be very immunosuppressive, enhancing the risk for invasive mold infections. Although the numbers illustrated in the slide are not statistically different, there may be a signal in these observations regarding the magnitude of this risk.

These observations are not sufficient to warrant consideration of routine deployment of mold-active prophylaxis; however, they do reinforce the need for more robust assessment of the actual event rates for invasive fungal infection among adult ALL patients receiving dose-intensive chemotherapy regimens.


There have been a number of antifungal prophylaxis studies published over the years. There are a number of lessons to be gleaned from the systematic reviews of these experiences. First, the risk for invasive fungal infection is not uniform across all patient groups, and the risks are discontinuous over time. For example, the risk for invasive fungal infection during remission induction in AML is higher than that associated with post-remission consolidation. Accordingly, antifungal prophylaxis may be appropriate for the former but not the latter situation. Second, clinical trials have demonstrated that relevant event rates such as proven and probable invasive fungal infection are reducible by chemoprophylaxis strategies.

Third, the efficacy of chemoprophylaxis also varies with the event rate. The lowest event rate for invasive fungal infection among control patients in comparative clinical trials appears to be of the order of 5 to 6%. The protective benefit of antifungal prophylaxis applied to a population where the baseline event rate is below this may be negligible, or at least not be detectable. It may not be worth our while to apply an antifungal prophylactic strategy in that patient population, given the expense of the drug, the toxicities of the drug, and the increasing concerns over antifungal drug resistance.

Fourth, efficacy varies with product and the pathogen. For example, an anti-aspergillosis prophylaxis strategy based upon fluconazole would be expected to fail based upon the lack of activity of this agent against Aspergillus spp. Accordingly, the clinician must choose the antifungal prophylaxis agent on the basis of the pathogen to be targeted and the antifungal activity of the agent. Fifth, the choice of formulation may impact upon antifungal efficacy. For example, systematic review of clinical trials evaluating the role of itraconazole to prevent invasive aspergillosis demonstrated relative lack of efficacy of the oral capsules compared with the oral solution. Sixth, efficacy varies with the dose of antifungal agent. Experience has suggested that lower doses may result in lower drug exposures and increased risk for treatment failures and breakthrough fungal infection.

Lastly, the optimal duration of prophylaxis remains unclear. Many pundits argue that antifungal therapy should continue until the end-of-risk. End-of-risk may be defined by recovery from myelosuppression, as in the circumstances of AML therapy-induced neutropenia, or by the discontinuance of immunosuppressive therapy as in the case of allogeneic stem cell transplant recipients treated successfully for acute graft-versus-host disease. The optimal duration for prophylaxis in adult ALL, where intensive treatments may last 8 or more months, remains undefined.


This is an illustrated summary of the circumstances in which published guidelines suggest yeast and mold prophylaxis should be considered. The green and red faces indicate recommendations for and against antifungal prophylaxis, respectively. The yellow face indicates insufficient evidence to support a recommendation (clinical equipoise). Yeast prophylaxis is not recommended in patients undergoing primary treatment for solid tumors or lymphoma. The use of purine analogue therapy enhances the immunosuppressive effect of chemotherapy in lymphoreticular malignancies; however, it remains unclear whether this effect translates into an enhanced risk for invasive fungal infection. There is evidence supporting yeast prophylaxis in the setting of primary and salvage induction therapy in AML (AI), but not post-remission consolidation. Similarly, the duration of the myelosuppression-driven risk for invasive candidiasis is sufficiently short among stem-cell autograft recipients, particularly where hematopoietic growth factors are being employed to hasten engraftment, such that anti-yeast prophylaxis is not recommended. There may be an argument for such prophylaxis among stem cell autograft recipients receiving myeloablative conditioning regimens without hematopoietic growth factor support and who are expected to have grade 3-4 mucositis. Yeast prophylaxis may be considered for stem-cell allograft recipients during the pre-engraftment phase post transplant and during the post-engraftment phase that is punctuated by graft-versus-host disease.


Mold-active prophylaxis is not recommended for patients receiving cyclical cytotoxic chemotherapy for solid tumors or lymphoma. There is a suggestion from case series that the risk of invasive mold infection among purine analogue recipients (eg, patients with advanced chronic lymphocytic leukemia) may be sufficiently high to warrant consideration of anti-mold prophylaxis. The evidence base to support this is not robust, however (CII). AML patients undergoing primary or secondary remission-induction may benefit from anti-mold prophylaxis if they are being treated outside a high-efficiency particulate air-filtered protected environment (AI). As for the considerations for yeast prophylaxis after post-remission consolidation, mold-active prophylaxis is not recommended for consolidation in AML; nor is it recommended in the setting of stem cell autografting. Although many clinicians prescribe anti-mold prophylaxis in the pre-engraftment setting post stem cell allografting, there is little clinical trial–based evidence to support this practice. It seems to be largely extrapolated from the AML induction experience. Lastly, mold-active prophylaxis is recommended in the circumstance of graft-versus-host disease post stem cell allografting.

This can be modified if your institution event rate is unacceptably high, that is above 8% or 9%; then that would justify reconsidering some of these guidelines.


Let me address pre-emptive therapy for just for a few moments. There are a number of studies that have tried to address how we should execute this strategy using the diagnostic technologies that we have available.

An early retrospective trial, a single arm trial from France examined the role of serial high resolution CT screening. Another nonrandomized study in AML patients, published by Johan Maertens, demonstrated a 78% reduction in the prescription of empirical anti-fungal therapy through serial serum galactomannan testing. A randomized-controlled German trial of serial serum Aspergillus PCR in stem cell transplant recipients, observed 56% strategy-driven increase in the use of empirical antifungal therapy, but no impact upon all-cause mortality.

Another randomized-controlled French study, from Cordonnier and colleagues, in acute leukemia and stem cell autograft patients using clinical signs and symptoms along with a serum galactomannan, observed a 36% reduction in the use of empirical amphotericin B therapy and an increase in the IFI detection rate, but no strategy-driven impart upon all-cause mortality.

A Swedish study examined a once-weekly pan-fungal PCR-based pre-emptive strategy for invasive fungal infection among reduced-intensity conditioned (RIC) stem cell allograft recipients during the first 100 post-transplant days. No impact of the monitoring strategy was detected before day 100. Most of the cases of IFI occurred after day 100, consistent with the period of greatest risk among RIC-conditioned allograft recipients.

Lastly, a randomized-controlled multicenter study in acute leukemia and stem-cell transplant recipients in Australia examined the impact of serial screening using both serial galactomannan and PCR-based assays. There was a strategy-driven 53% reduction in the use of empirical antifungal therapy but no impact on directed antifungal therapy or all-cause mortality.

These studies illustrate proof of principle that diagnostics-driven testing for biological markers of invasive fungal infection can impact on the empirical use of antifungal therapy and also reduce the time to treatment.


What do our guidelines say about this? The IDSA guidelines saw pre-emptive as a logical extension to empirical antifungal therapy, but no recommendations could be offered on the basis of the available information at the time. The Italian guidelines recommend a “surveillance strategy and/or an active diagnostic approach.” The Canadian guidelines could not make a recommendation based upon the information available in 2009-2010. The 2009 European Conference on Infections in Leukaemia–3 was unable to make any recommendation because of a lack of standardized definitions. The 2010 IDSA guidelines on the use of antimicrobial agents in neutropenic patients suggested that a diagnostics-driven pre-emptive approach may become an acceptable alternative to empirical antifungal therapy strategies, but couldn’t make a firm recommendation at that time.


There are a number of risk factors that have been identified for invasive fungal infection among patients undergoing hematopoietic stem cell transplantation. Many of these are listed on the left hand side of the slide. Some are dependent upon one another, and others are independent predictors of IFI. Some may even interact with one another with synergistic or additive effects.

After review, some of these factors such as steroid-refractory graft-versus-host disease, cytomegalovirus DNAemia, and graft failure have been prominent in the literature as variables linked to enhanced risk for IFI.


The above figure is offered as a model of how these predictor variables might be used to decide which patients might be candidates for mold-active chemoprophylaxis or a diagnostics-driven pre-emptive strategy.

For example, the onset of GvHD among stem cell allograft recipients is a known risk factor for IFI (see the pathway at the top of the figure). Among the sub-group who prove to have steroid-refractory GvHD and for whom augmented immunosuppression is required, the risk for IFI may be even greater. Such patients may be candidates for a mold-active prophylaxis approach, whereas a diagnostics-driven pre-emptive monitoring approach may be appropriate for those whose GvHD responds to primary steroid-based therapy.

Among patients who shed Cytomegalovirus (CMV, a known risk for IFI in stem cell transplant recipients – see the middle pathway in the figure), a mold-active prophylaxis approach might be considered for those who also develop GvHD, but a pre-emptive monitoring approach may be considered for those who remain free of GvHD.

Similarly, among those transplant recipients who develop graft failure, a mold-active prophylaxis approach may be considered if there is concomitant CMV DNAemia, and pre-emptive monitoring may be considered for those without CMV DNAemia.

These possibilities would limit the exposure of mold-active triazole prophylaxis to very selected sub-groups of patients at risk for IFI. Such strategic management pathways will require validation in prospective trials before they can be accepted and deployed for routine use, however.


Empirical antifungal therapies are administered to patients at risk for IFI and who have a febrile illness that could be driven by an invasive fungal infection, but for whom there is no mycological and very limited clinical evidence to support an IFI diagnosis. A persistent neutropenic fever (PNF) syndrome occurring in the setting of broad-spectrum antibacterial therapy is an example of such a circumstance (the case definition for which appears on the slide that follows).



The approaches clinicians have employed for the PNF have varied depending upon the clinical circumstances (see the above figure). For example, a survey of European clinicians’ antifungal prescriptions demonstrated a preference for conventional amphotericin B deoxycholate for empirical antifungal therapy for the PNF syndrome in the setting of unexplained fever during acute leukemia induction or post-remission consolidation. However, liposomal amphotericin B was preferred in the setting of allogeneic stem cell transplantation, or in the setting of PNF with clinical instability. A mold-active azole, voriconazole, was preferred if the PNF syndrome was accompanied by pulmonary infiltrates. An echinocandin or fluconazole was preferred in the setting of neutropenic enterocolitis.


Based upon published experience (see the above figure), the PNF syndrome may be seen in approximately one-in-five to one-in-two neutropenic subjects. Most (96%) of these neutropenic fevers remain unexplained (that is, no focus and no pathogen identified). Of these febrile events, a smaller proportion of PNF syndromes (2-8%) may be associated with breakthrough IFI, whilst the patient continues to receive the empirical antifungal agent. These breakthrough IFIs are divided between invasive candidiasis and an invasive mold infection. A small proportion (~4%) of PNF syndromes may be explained by a baseline IFI, either a mold or an invasive yeast infection. Treatment of these events must take into consideration the antifungal agents received by the patient as either antifungal prophylaxis or as empirical antifungal therapy. Such patients will be treated until response or until a change is required primarily for treatment failure or for toxicity.


The choice of prophylactic antifungal agent influences the choice of the class of antifungal agent to be used for empirical or directed therapy. For example, breakthrough IFIs in patients receiving fluconazole-based anti-fungal prophylaxis may be salvaged using an extended-spectrum mold-active azole (ESA) such as voriconazole, or an echinocandin such as caspofungin, or a polyene such as liposomal amphotericin B. Given the similarities in antifungal spectrum among ESAs and echinocandins, salvage therapy for breakthrough IFI for patients receiving ESAs or echinocandins as prophylaxis is limited to liposomal amphotericin. However, salvage antifungal therapy for systemic amphotericin-based prophylaxis failures is limited to ESAs or echinocandins, the similarities of activity spectra notwithstanding.


Directed antifungal therapy is based upon a hierarchical classification of diagnostic robustness; that is, possible, probable, or proven IFI. The clinician must consider the syndrome and the possible mycological etiologies, the context of the underlying cancer and treatment for that cancer, prior exposure to azoles and the possibility of drug resistance, the role of monotherapy and combination antifungal therapy, the role of risk factors such as the presence of central venous access devices that may be amenable to modification, the duration of antifungal treatment required for a given syndrome, and the role of therapeutic drug monitoring for azole therapy to optimize efficacy and minimize toxicity.


Voriconazole has become the accepted standard of practice for the treatment of invasive aspergillosis in high-risk patients. Based on the seminal study of Herbrecht and colleagues in 2002 comparing voriconazole to the then-standard, amphotericin B deoxycholate, a significant clinical response and survival advantage was demonstrated among the voriconazole recipients. Moreover, the treatment effect was observed among different diagnostic sub-groups of patients (see the above figure for the relative responses among acute leukemic neutropenic patients and stem-cell transplant patients). It was noted that response rates among stem-cell transplant recipients was significantly lower than among other risk groups, suggesting that more work was needed to improve these outcomes for this group of patients.


Newer agents such as isavuconazole have been studied for invasive aspergillosis. The results of a recent comparative trial with voriconazole have suggested similar efficacy but a modest toxicity advantage for isavuconazole.


A recent blinded, randomized-controlled trial by Marr and colleagues (see the above figure) of combination antifungal therapy for invasive aspergillosis in stem-cell transplant recipients and patients under treatment for hematological malignancy demonstrated a survival advantage for a combination of voriconazole and an echinocandin, anidulafungin, among patients with galactomannan-based probable invasive aspergillosis who had a moderate burden of infection (as measured by serum galactomannan – see figure below).



The published guidelines for invasive aspergillosis (IA) do not support combination antifungal therapy for the primary treatment of IA. There is more support for the use of combination strategies in salvage IA therapy.


The issue of extended-spectrum azole resistance among Aspergillus spp. has been raised by several investigators. The above map from the group in Nijmegen, the Netherlands, demonstrating the wide distribution of resistant strains (in the shaded areas) that have been identified. This phenomenon is raised as a cautionary note.

Let me just summarize what I have learned.

  • Antifungal prophylaxis strategies may have a protective treatment effect against molds and yeasts; but, ...
  • It remains unclear - who gets the most benefit and the best value for the dollar spent?
  • Whilst the reported experience with diagnostics-based pre-emptive therapy looks promising, we don’t yet know the optimal drivers. In some clinical circumstances this strategy may replace antifungal prophylaxis.
  • Persistent neutropenic fever–driven empirical antifungal therapy has limited utility. It adds cost and certainly over-treats and adds to the risk of toxicity.
  • Based on the available evidence, voriconazole today represents the monotherapy of choice for proven/probable invasive aspergillosis. The fungal disease burden also influences the outcome and prognosis for patients.
  • Combination antifungal therapy for invasive aspergillosis infections is experimental but looks very promising. The one study that we have discussed is promising but in a very distinct subgroup of patients for whom the diagnosis was established by the use of serum galactomannan.
  • Azole-based mold-active prophylaxis will likely preclude the use of azole-based combination antifungal therapy. It is unclear whether the efficacy of azole:liposomal amphotericin B combination therapy is equivalent to azole:echinocandin therapy.
  • The ever-present threat of resistance among fungi may represent a limiting factor in treatment success. Increasing antifungal resistance may threaten the effectiveness of the strategies that have recently been developed. Antifungal stewardship in the form of a disciplined approach to the deployment of these agents may limit, at least in part, the impact of this phenomenon.

SECTION 5: PANEL DISCUSSION

QUESTION:

To the panel, can you comment on some of the major flaws in the Gomes study? Namely the use of IFI incidence per prophylactic days (where echinocandins were only used in the first 30 days, the highest risk period and likely) in the patients receiving hepatotoxic chemo, ie, clofarabine. Fluconazole actually had the lowest rate of IFIs per their definition. Basically how comfortable are you in using echinocandin prophylaxis and treatment in various setting?

ANSWER:

JAMES LEWIS, II, PharmD:

I think the author of the question really hits a lot of the nails on the head with regards to the limitations of that study. And I think the fact that it’s a single-center experience with a very high attack rate are 2 other limitations to the data set that I would certainly add.

That being said, though, the fact of the matter is that there's still something of a signal there. And it is one of those things where I will certainly use the echinocandin in a prophylaxis setting when there is a good reason not to be using the azole.

And I think what that data says to me is that I will do that, but I will try to get them back to the azole as quickly as I can for a more prolonged prophylaxis-type setting. Again it’s anecdotal single-center experience, but we certainly seem to see more breakthroughs with echinocandins, as well. Again, limitation, single-center, anecdotal response to the question.

But it does give me a little bit of pause. So I think long story short, I would still do it if absolutely necessary in a situation where the azole therapy is not optimal, for whatever reason, but will try to get back to an azole as quickly as I can.

QUESTION FROM AUDIENCE MEMBER:

As I go through the slides, I see one of the highest incidence of fungal infection is in patients with acute lymphoblastic leukemia. And one of the problems in prophylaxis over there is if you use azole, there is an interaction with vincristine, which is the mainstay of therapy – and there is an interaction between vincristine and azoles. And if you use something like micafungin, it’s an intravenous therapy and you cannot do this in the outpatient setting.

So let me ask, in each of your institutions, how do you deal with that? And sometimes the toxicity is really bad if you're using azoles that you have to stop their ALL therapy. And that has affected their outcomes.

ANSWER:

MATTHEW LUNNING, DO:

That’s an excellent question. In fact, that was something that we spent I think a good 10 minutes on before the actual symposium discussing about it. Because internally in our own ALL use, whether it’s with HyperCVAD in ALL, or trying to use some of the earlier BFM regimens and AYAs, we struggle with this concept. And I don't know that there's definitely a right answer.

ALISON FREIFELD, MD:

I don’t think there is a very good answer. And I will say that, for example, the NCCN guidelines suggest an echinocandin. And I know that’s very burdensome for the patient. But we also try to avoid the azoles in that situation. There's not a great answer for that because of the severe neurotoxicity.

MATTHEW LUNNING, DO:

And I think that sometimes, if you will, people try to use echinocandins as an outpatient in a different dosing and schedule. I think it’s 300 Monday, Wednesday, Friday is something that I have seen be done.

But where is the data for that? Well, it’s often single institution; this is how we try to do it, rather than, “There's a paper to be referenced.”

JAMES LEWIS, II, PharmD:

But again I think to follow up, that was exactly the point I was going to make. There's a very nice supplement that was—I believe it was sponsored by Astellas—that was in Clinical Infectious Diseases not too long ago.

Really, when you look at echinocandin half-life and you look at the fact that those are concentration-dependent drugs, or at least they appear to be in vitro, some of these extended dosing interval uses of the echinocandins I think really do make a lot of sense pharmacologically. And while I understand that there are definite limitations, the currently available data, I think that again given the risk of the azoles with the vincristines and whatnot of the world, that those types of regimens make a lot of sense.

ERIC BOW, MD:

Again I think what you've done is identified an unmet need for research. And again, many individuals—at least in the Canadian centers—have chosen to go back to the old school of using amphotericin B in intermittent dosing. And again, I know of no specific data in adult ALL using HyperCVAD-like regimens that gives us confidence in that strategy. I’ll have to argue, just as an aside, I don't know that we actually know what the event rates for invasive fungal disease—and mold disease in particular—is in that patient population as an ambulatory type of population.

AUDIENCE MEMBER:

At our own center, we have been doing it for the first cycle they are usually inpatient, so we use an echinocandin. But for the subsequent cycles, for the Part B, we go to an azole. And for the third cycle, we try to start an azole 48 hours after vincristine and give a break. But I'm not sure...

ALISON FREIFELD, MD:

That’s reasonable.

MATTHEW LUNNING, DO:

And I think the risk probably changes as you get further along in ALL management obviously. Upfront they're at much higher risk because you can never tell how long they’ve been immunosuppressed functionally from their disease, leading into cycles 1a and 1b, or induction and intensification from that standpoint. Johan, did you have a comment?

JOHAN MAERTENS, MD:

Yeah, I wanted to comment on Eric’s suggestion of using a polyene. You have to know that Gilead just did a study looking at adult ALL patients, where they randomized them between either placebo or a liposomal amphotericin B given twice weekly at 5 mg/kg during the first cycle.

And it’s quite a large study, it’s around 300 patients. The incidence of invasive fungal infection was much higher than what we saw in the Italian study, that was 7.5%. In the Gilead study, it’s 12%. So it’s really almost the same size in what we see in the AML population.

But the key message of the study was that there was no difference between the placebo arm and the liposomal amphotericin B arm. Now that may be because the dose was too low, or maybe you have to give it 3 times a week instead of twice. That we don’t know.

But if you look at the study results, there was no real difference between placebo or liposomal amphotericin B during the first cycle of ALL treatment.

ALISON FREIFELD, MD:

And I think that’s a good point. And also to follow up on what Matt is saying, if we use AML as the paradigm, what I showed was that the incidence of invasive fungal disease is maybe 3% during consolidation cycle.

So as you control the underlying disease, your rates of invasive fungal infection are markedly declined. So the first cycle is the one obviously that—especially as you're underscoring—probably makes the most sense in which to do the most intensive prophylaxis.

QUESTION: This will be for Johan to give you some time to think. How often do you see false positive makers, ie (1→3)-β-D-glucan when patients are dialysis?

ANSWER:

JOHAN MAERTENS, MD:

Oh, a lot. If you have a patient on dialysis and (1→3)-β-D-glucan is quite difficult to use. So let's say that most of the false positives we see in the ICU setting, patients receiving hemoglobins, lots of blood products, being on dialysis, being post surgery—it’s a difficult assay to handle in the ICU setting or in the critical outpatient population.

We don’t see that many false positives in let's say the neutropenic hematology population. But as you mentioned on dialysis, it really is a problem.

QUESTION FROM AUDIENCE:

Hi, John Wilson from Mayo Clinic. And we've been wrestling with the use of isavuconazole in our hematology patients, whether prophylactically or therapeutically and looking at the potential, just because of cost issues and looking at the potential role for patients that have underlying QTc prolongation or drug interactions in that capacity.

But we're also looking at the long half-life of isavuconazole and the potential use of antineoplastics with CYP3A4 drug interactions in that regard, and given that long terminal half-life I think of 130-140 hours, should we potentially be looking at a washout period of stopping isavuconazole, looking at another potential anti-mold agent for a period of time before embarking on other agents with CYP3A4 drug interactions, or additional antineoplastic chemotherapy options that go down that route?

ANSWER:

JAMES LEWIS, II, PharmD:

I sure as heck would. I completely agree with you that that long half-life, while it’s of benefit from a therapeutic and ease of dosing standpoint, in the situations you're describing, it really represents a challenge. That inhibition of the P-450 system does not go away for at least the first 2 to 3 half-lives. And it’s probably even a little bit further out than that. So I really agree with your comments and your concerns.

References

  • Allen KC, Sanchez CJ Jr, Niece KL, Wenke JC, Akers KS. Voriconazole enhances the osteogenic activity of human osteoblasts in vitro through a fluoride-independent mechanism. Antimicrob Agents Chemother. 2015;59:7205-7213.
  • Ananda-Rajah MR, Grigg A, Downey MT, et al. Comparative clinical effectiveness of prophylactic voriconazole/posaconazole to fluconazole/itraconazole in patients with acute myeloid leukemia/myelodysplastic syndrome undergoing cytotoxic chemotherapy over a 12-year period. Haematologica. 2012;97:459-463.
  • Andes D, Pascual A, Marchetti O. Antifungal therapeutic drug monitoring: established and emerging indications. Antimicrob Agents Chemother. 2009;53:24-34.
  • Arvanitis M, Anagnostou T, Mylonakis E. Galactomannan and polymerase chain reaction-based screening for invasive aspergillosis among high-risk hematology patients: a diagnostic meta-analysis. Clin Infect Dis. 2015;61:1263-1272.
  • Arvanitis M, Ziakas PD, Zacharioudakis IM, Zervou FN, Caliendo AM, Mylonakis E. PCR in diagnosis of invasive aspergillosis: a meta-analysis of diagnostic performance. J Clin Microbiol. 2014;52:3731-3742.
  • Astellas Anti-Infective Drugs Advisory Committee. Briefing Document: Isavuconazonium Invasive Aspergillosis and Invasive Mucormycosis. Northbrook, IL: Astellas Pharma US, Inc.; 2014. http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ AntiInfectiveDrugsAdvisoryCommittee/UCM430748.pdf. Accessed 10/15.
  • Baddley JW, Andes DR, Marr KA, et al. Factors associated with mortality in transplant patients with invasive aspergillosis. Clin Infect Dis. 2010;50:1559-1567.
  • Barnes RA, Stocking K, Bowden S, Poynton MH, White PL. Prevention and diagnosis of invasive fungal disease in high-risk patients within an integrative care pathway. J Infect. 2013;67:206-214.
  • Blennow O, Remberger M, Klingspor L, et al. Randomized PCR-based therapy and risk factors for invasive fungal infection following reduced-intensity conditioning and hematopoietic SCT. Bone Marrow Transplant. 2010;45:1710-1718.
  • Bow EJ, Evans G, Fuller J, et al. Canadian clinical practice guidelines for invasive candidiasis in adults. Can J Infect Dis Med Microbiol. 2010;21:e122-e150.
  • Brüggemann RJ, Alffenaar JW, Blijlevens NM, et al. Clinical relevance of the pharmacokinetic interactions of azole antifungal drugs with other coadministered agents. Clin Infect Dis. 2009;48:1441-1458.
  • Bueid A, Howard SJ, Moore CB, et al. Azole antifungal resistance in Aspergillus fumigatus: 2008 and 2009. J Antimicrob Chemother. 2010;65:2116-2118.
  • Caillot D, Casasnovas O, Bernard A, et al. Improved management of invasive pulmonary aspergillosis in neutropenic patients using early thoracic computed tomographic scan and surgery. J Clin Oncol. 1997;15:139-147.
  • Caira M, Candoni A, Verga L, et al; SEIFEM Group (Sorveglianza Epidemiologica Infezioni Fungine in Emopatie Maligne). Pre-chemotherapy risk factors for invasive fungal diseases: prospective analysis of 1,192 patients with newly diagnosed acute myeloid leukemia (SEIFEM 2010-a multicenter study). Haematologica. 2015;100:284-292.
  • Cordonnier C, Pautas C, Maury S, et al. Empirical versus preemptive antifungal therapy for high-risk, febrile, neutropenic patients: a randomized, controlled trial. Clin Infect Dis. 2009;48:1042-1051.
  • Cornely O, Maertens J, Bresnik M, et al; AmBiLoad Trial Study Group. Liposomal amphotericin B as initial therapy for invasive mold infection: a randomized trial comparing a high-loading dose regimen with standard dosing (AmBiLoad trial). Clin Infect Dis. 2007;44:1289-1297.
  • Cornely OA, Maertens J, Winston DJ, et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med. 2007;356:348-359.
  • Cresemba prescribing information. Northbrook, IL: Astellas Pharma; 2015.
  • De Pauw B, Walsh TJ, Donnelly JP, et al; European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group; National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Revised definitions of invasive fungal disease from the European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin Infect Dis. 2008;46:1813-1821.
  • Diflucan prescribing information. New York, NY: Pfizer; 2014.
  • Doan TN, Kirkpatrick CM, Walker P, et al. Primary antifungal prophylaxis in adult patients with acute lymphoblastic leukaemia: a multicentre audit. J Antimicrob Chemother. 2015;71:497-505.
  • Donnelly JP, Leeflang MM. Galactomannan detection and diagnosis of invasive aspergillosis. Clin Infect Dis. 2010;50:1070-1071.
  • Duarte RF, Sánchez-Ortega I, Cuesta I, et al. Serum galactomannan-based early detection of invasive aspergillosis in hematology patients receiving effective antimold prophylaxis. Clin Infect Dis. 2014;59:1696-1702.
  • Flowers CR, Seidenfeld J, Bow EJ, et al. Antimicrobial prophylaxis and outpatient management of fever and neutropenia in adults treated for malignancy: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2013;31:794-810.
  • Freifeld AG, Bow EJ, Sepkowitz KA, et al; Infectious Diseases Society of America. Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the Infectious Diseases Society of America. Clin Infect Dis. 2011;52:e56-e93.
  • Girmenia C, Barosi G, Aversa F, et al. Prophylaxis and treatment of invasive fungal diseases in allogeneic stem cell transplantation: results of a consensus process by Gruppo Italiano Trapianto di Midollo Osseo (GITMO). Clin Infect Dis. 2009;49:1226-1236.
  • Girmenia C, Raiola AM, Piciocchi A, et al. Incidence and outcome of invasive fungal diseases after allogeneic stem cell transplantation: a prospective study of the Gruppo Italiano Trapianto Midollo Osseo (GITMO). Biol Blood Marrow Transplant. 2014;20:872-880.
  • Glasmacher A, Cornely O, Ullmann AJ, et al; Itraconazole Research Group of Germany. An open-label randomized trial comparing itraconazole oral solution with fluconazole oral solution for primary prophylaxis of fungal infections in patients with haematological malignancy and profound neutropenia. J Antimicrob Chemother. 2006;57:317-325.
  • Gomes MZ, Jiang Y, Mulanovich VE, Lewis RE, Kontoyiannis DP. Effectiveness of primary anti-Aspergillus prophylaxis during remission induction chemotherapy of acute myeloid leukemia. Antimicrob Agents Chemother. 2014;58:2775-2780.
  • Goyal RK. Voriconazole-associated phototoxic dermatoses and skin cancer. Expert Rev Anti Infect Ther. 2015;13:1537-1546.
  • Harousseau JL, Dekker AW, Stamatoullas-Bastard A, et al. Itraconazole oral solution for primary prophylaxis of fungal infections in patients with hematological malignancy and profound neutropenia: a randomized, double-blind, double-placebo, multicenter trial comparing itraconazole and amphotericin B. Antimicrob Agents Chemother. 2000;44:1887-1893.
  • He S, Hang JP, Zhang L, Wang F, Zhang DC, Gong FH. A systematic review and meta-analysis of diagnostic accuracy of serum 1,3-β-D-glucan for invasive fungal infection: focus on cutoff levels. J Microbiol Immunol Infect. 2015;48:351-361.
  • Hebart H, Klingspor L, Klingebiel T, et al. A prospective randomized controlled trial comparing PCR-based and empirical treatment with liposomal amphotericin B in patients after allo-SCT. Bone Marrow Transplant. 2009;43:553-561.
  • Heng SC, Morrissey O, Chen SC, et al. Utility of bronchoalveolar lavage fluid galactomannan alone or in combination with PCR for the diagnosis of invasive aspergillosis in adult hematology patients: a systematic review and meta-analysis. Crit Rev Microbiol. 2015;41:124-134.
  • Herbrecht R, Flückinger U, Gachot B, Ribaud, Thiebaut A, Cordonnier C. Treatment of invasive Candida and invasive Aspergillus infections in adult haematological patients. Eur J Cancer Suppl. 2007;5:49-59.
  • Herbrecht R, Denning DW, Patterson TF, et al; Invasive Fungal Infections Group of the European Organisation for Research and Treatment of Cancer and the Global Aspergillus Study Group. Voriconazole versus amphotericin B for primary therapy of invasive aspergillosis. N Engl J Med. 2002;347:408-415.
  • Hou TY, Wang SH, Liang SX, Jiang WX, Luo DD, Huang DH. The screening performance of serum 1,3-beta-D-glucan in patients with invasive fungal diseases: a meta-analysis of prospective cohort studies. PLoS One. 2015;10:e0131602.
  • Karageorgopoulos DE, Vouloumanou EK, Ntziora F, Michalopoulos A, Rafailidis PI, Falagas ME. β-D-glucan assay for the diagnosis of invasive fungal infections: a meta-analysis. Clin Infect Dis. 2011;52:750-770.
  • Kontoyiannis DP. Invasive mycoses: strategies for effective management. Am J Med. 2012;125(1 Suppl):S25-S38.
  • Kontoyiannis DP, Marr KA, Park BJ, 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;50:1091-1100.
  • Krishna G, Ma L, Martinho M, Preston RA, O'Mara E. A new solid oral tablet formulation of posaconazole: a randomized clinical trial to investigate rising single- and multiple-dose pharmacokinetics and safety in healthy volunteers. J Antimicrob Chemother. 2012;67:2725-2730.
  • Lamoth F, Cruciani M, Mengoli C, et al; Third European Conference on Infections in Leukemia (ECIL-3). β-Glucan antigenemia assay for the diagnosis of invasive fungal infections in patients with hematological malignancies: a systematic review and meta-analysis of cohort studies from the Third European Conference on Infections in Leukemia (ECIL-3). Clin Infect Dis. 2012;54:633-643.
  • Leeflang MM, Debets-Ossenkopp YJ, Visser CE, et al. Galactomannan detection for invasive aspergillosis in immunocompromized patients. Cochrane Database Syst Rev. 2008;(4):CD007394.
  • Maertens J, Marchetti O, Herbrecht R, et al; Third European Conference on Infections in Leukemia. European guidelines for antifungal management in leukemia and hematopoietic stem cell transplant recipients: summary of the ECIL 3--2009 update. Bone Marrow Transplant. 2011;46:709-718.
  • Maertens J, Theunissen K, Verhoef G, et al. Galactomannan and computed tomography-based preemptive antifungal therapy in neutropenic patients at high risk for invasive fungal infection: a prospective feasibility study. Clin Infect Dis. 2005;41:1242-1250.
  • Maertens JA, Nucci M, Donnelly JP. The role of antifungal treatment in hematology. Haematologica. 2012;97:325-327.
  • Maertens JA, Raad II, Marr KA, et al. Isavuconazole versus voriconazole for primary treatment of invasive mould disease caused by Aspergillus and other filamentous fungi (SECURE): a phase 3, randomised-controlled, non-inferiority trial. Lancet. 2016;387:760-769.
  • Malani AN, Kerr L, Obear J, Singal B, Kauffman CA. Alopecia and nail changes associated with voriconazole therapy. Clin Infect Dis. 2014;59:e61-e65.
  • Marchetti O, Lamoth F, Mikulska M, Viscoli C, Verweij P, Bretagne S; European Conference on Infections in Leukemia (ECIL) Laboratory Working Groups. ECIL recommendations for the use of biological markers for the diagnosis of invasive fungal diseases in leukemic patients and hematopoietic SCT recipients. Bone Marrow Transplant. 2012;47:846-854.
  • Marchetti O, Cordonnier C, Calandra T. Empirical antifungal therapy in neutropaenic cancer patients with persistent fever. Eur J Cancer Suppl. 2007;5:32-42.
  • Marks DI, Pagliuca A, Kibbler CC, et al. Voriconazole versus itraconazole for antifungal prophylaxis following allogeneic haematopoietic stem-cell transplantation. Br J Haematol. 2011;155:318-327.
  • Marr KA, Crippa F, Leisenring W, et al. Itraconazole versus fluconazole for prevention of fungal infections in patients receiving allogeneic stem cell transplants. Blood. 2004;103:1527-1533.
  • Marr KA, Schlamm HT, Herbrecht R, et al. Combination antifungal therapy for invasive aspergillosis: a randomized trial. Ann Intern Med. 2015;162:81-89.
  • Marr KA, Laverdiere M, Gugel A, Leisenring W. Antifungal therapy decreases sensitivity of the Aspergillus galactomannan enzyme immunoassay. Clin Infect Dis. 2005;40:1762-1769.
  • Mengoli C, Cruciani M, Barnes RA, Loeffler J, Donnelly JP. Use of PCR for diagnosis of invasive aspergillosis: systematic review and meta-analysis. Lancet Infect Dis. 2009;9:89-96.
  • Mennink-Kersten MASH, Donnelly JP, Verweij PE. Detection of circulating galactomannan for the diagnosis and management of invasive aspergillosis. Lancet Infect Dis. 2004;4:349-357.
  • Miceli HM, Maertens J. Role of non-culture-based tests, with an emphasis on galactomannan testing for the diagnosis of invasive aspergillosis. Semin Respir Crit Care Med. 2015;36:650-661.
  • Moon WJ, Scheller EL, Suneja A, et al. Plasma fluoride level as a predictor of voriconazole-induced periostitis in patients with skeletal pain. Clin Infect Dis. 2014;59:1237-1245.
  • Morrissey CO, Chen SC, Sorrell TC, et al; Australasian Leukaemia Lymphoma Group and the Australia and New Zealand Mycology Interest Group. Galactomannan and PCR versus culture and histology for directing use of antifungal treatment for invasive aspergillosis in high-risk haematology patients: a randomised controlled trial. Lancet Infect Dis. 2013;13:519-528.
  • National Comprehensive Cancer Network. Prevention and Treatment of Cancer-Related Infections. NCCN Guidelines Version 1.2016. Fort Washington, PA: National Comprehensive Cancer Network, Inc.; 2016.
  • Neofytos D, Lu K, Hatfield-Seung A, et al. Epidemiology, outcomes, and risk factors of invasive fungal infections in adult patients with acute myelogenous leukemia after induction chemotherapy. Diagn Microbiol Infect Dis. 2013;75:144-149.
  • Noxafil prescribing information. Whitehouse Station, NJ: Merck; 2015.
  • Obayashi T, Yoshida M, Mori T, et al. Plasma (1-->3)-beta-D-glucan measurement in diagnosis of invasive deep mycosis and fungal febrile episodes. Lancet. 1995;345:17-20.
  • Odabasi Z, Paetznick VL, Rodriguez JR, Chen E, McGinnis MR, Ostrosky-Zeichner L. Differences in beta-glucan levels in culture supernatants of a variety of fungi. Med Mycol. 2006;44:267-272.
  • Pagano L, Caira M, Candoni A, et al. The epidemiology of fungal infections in patients with hematologic malignancies: the SEIFEM-2004 study. Haematologica. 2006;91:1068-1075.
  • Pagano L, Caira M, Candoni A, et al. Invasive aspergillosis in patients with acute myeloid leukemia: a SEIFEM-2008 registry study. Haematologica. 2010;95:644-650.
  • Pascual A, Csajka C, Buclin T, et al. Challenging recommended oral and intravenous voriconazole doses for improved efficacy and safety: population pharmacokinetics-based analysis of adult patients with invasive fungal infections. Clin Infect Dis. 2012;55:381-390.
  • Perfect JR, Hachem R, Wingard JR. Update on epidemiology of and preventive strategies for invasive fungal infections in cancer patients. Clin Infect Dis. 2014;59(suppl 5):S352-S355.
  • Pfeiffer CD, Fine JP, Safdar N. Diagnosis of invasive aspergillosis using a galactomannan assay: a meta-analysis. Clin Infect Dis. 2006;42:1417-1427.
  • Pham AN, Bubalo JS, Lewis JS 2nd. Comparison of posaconazole serum concentrations from haematological cancer patients on posaconazole tablet and oral suspension for treatment and prevention of invasive fungal infections. Mycoses. [Epub ahead of print]. 2016 Jan 6. doi: 10.1111/myc.12452.
  • Pini P, Bettua C, Orsi CF, et al. Evaluation of serum (1 → 3)-β-D-glucan clinical performance: kinetic assessment, comparison with galactomannan and evaluation of confounding factors. Infection. 2015 Oct 16. [Epub ahead of print].
  • Stynen D, Sarfati J, Goris A, et al. Rat monoclonal antibodies against Aspergillus galactomannan. Infect Immun. 1992;60:2237-2245.
  • Sulahian A, Porcher R, Bergeron A, et al. Use and limits of (1-3)-β-d-glucan assay (Fungitell), compared to galactomannan determination (Platelia Aspergillus), for diagnosis of invasive aspergillosis. J Clin Microbiol. 2014;52:2328-2333.
  • Thornton CR. Development of an immunochromatographic lateral-flow device for rapid serodiagnosis of invasive aspergillosis. Clin Vaccine Immunol. 2008;15:1095-1105.
  • Ullmann A, Lipton JH, Vesole DH, et al. Posaconazole or fluconazole for prophylaxis in severe graft-versus-host disease. N Engl J Med. 2007;356:335-347.
  • Van Leeuwen RW, van Gelder T, Mathijssen RH, Jansman FG. Drug-drug interactions with tyrosine-kinase inhibitors: a clinical perspective. Lancet Oncol. 2014;15:e315-e326.
  • Vega K, Kalkum M. Chitin, chitinase responses, and invasive fungal infections. Int J Microbiol. 2012;2012:920459.
  • Vermeulen E, Lagrou K, Verweij PE. Azole resistance in Aspergillus fumigatus: a growing public health concern. Curr Opin Infect Dis. 2013;26:493-500.
  • Vfend prescribing information. New York, NY: Pfizer; 2003.
  • Walsh TJ, Anaissie EJ, Denning DW, et al; Infectious Diseases Society of America. Treatment of aspergillosis: clinical practice guidelines of the Infectious Diseases Society of America. Clin Infect Dis. 2008;46:327-360.
  • Walsh TJ, Finberg RW, Arndt C, et al. Liposomal amphotericin B for empirical therapy in patients with persistent fever and neutropenia. National Institute of Allergy and Infectious Diseases Mycoses Study Group. N Engl J Med. 1999;340:764-771.
  • Walsh TJ, Pappas P, Winston DJ, et al. Voriconazole compared with liposomal amphotericin B for empirical antifungal therapy in patients with neutropenia and persistent fever. N Engl J Med. 2002;346:225-234.
  • Walsh TJ, Teppler H, Donowitz GR, et al. Caspofungin versus liposomal amphotericin B for empirical antifungal therapy in patients with persistent fever and neutropenia. N Engl J Med. 2004:351:1391-1402.
  • Weissinger F, Auner HW, Bertz H, et al. Antimicrobial therapy of febrile complications after high-dose chemotherapy and autologous hematopoietic stem cell transplantation—guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society of Hematology and Oncology (DGHO). Ann Hematol. 2012;91:1161-1174.
  • White PL, Perry MD, Loeffler J, et al; European Aspergillus PCR Initiative. Critical stages of extracting DNA from Aspergillus fumigatus in whole-blood specimens. J Clin Microbiol. 2010;48:3753-3755.
  • Williams K, Mansh M, Chin-Hong P, Singer J, Arron ST. Voriconazole-associated cutaneous malignancy: a literature review on photocarcinogenesis in organ transplant recipients. Clin Infect Dis. 2014;58:997-1002.
  • Wingard JR, Carter SL, Walsh TJ, et al; Blood and Marrow Transplant Clinical Trials Network. Randomized, double-blind trial of fluconazole versus voriconazole for prevention of invasive fungal infection after allogeneic hematopoietic cell transplantation. Blood. 2010;116:5111-5118.
  • Winston DJ, Maziarz RT, Chandrasekar PH, et al. Intravenous and oral itraconazole versus intravenous and oral fluconazole for long-term antifungal prophylaxis in allogeneic hematopoietic stem-cell transplant recipients: a multicenter, randomized trial. Ann Intern Med. 2003;138:705-713.
  • Wojenski DH, Bartoo GT, Merten JA, et al. Voriconazole exposure and the risk of cutaneous squamous cell carcinoma in allogeneic hematopoietic stem cell transplant patients. Transpl Infect Dis. 2015;17:250-258.
  • Zonios DI, Gea-Banacloche J, Childs R, Bennett JE. Hallucinations during voriconazole therapy. Clin Infect Dis. 2008;47:e7-e10.

Back to Top